1
|
Sun C, Zhou Z, Liu F, Li H, Liu Z. Combretastatin A4 phosphate encapsulated in hyaluronic acid nanoparticles is highly cytotoxic to oral squamous cell carcinoma. Arch Med Sci 2024; 20:1022-1028. [PMID: 39050147 PMCID: PMC11264095 DOI: 10.5114/aoms/189535] [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: 12/21/2023] [Accepted: 05/31/2024] [Indexed: 07/27/2024] Open
Abstract
Introduction To investigate the toxicity of combretastatin A4 phosphate (CA4P) hyaluronic acid (HA) gel nanoparticles (HA-CA4P-NPs) in OSCC (oral squamous cell carcinoma). Methods Toxicity was investigated using fluorescence microscopy, MTT assay, flow cytometry, and OSCC xenograft mouse models. Results Compared with CA4P, HA-CA4P-NPs generated nearly 10 times more fluorescence in OSCC cells. Cytotoxicity assays showed that HACA4P-NPs were more toxic to SCC-4 cells but not to HNECs. Remarkable necrosis was induced in SCC-4 cells after exposure to HA-CA4P-NPs, and related proteins were upregulated. Furthermore, HA-CA4P-NPs significantly reduced the tumour size. Conclusions HA-CA4P-NPs improved drug release and delivery, and increased cytotoxicity to cancer cells.
Collapse
Affiliation(s)
- Chuanxi Sun
- Department of Orthodontics, The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, Jiangxi Province, China
| | - Ziqi Zhou
- Department of Orthodontics, The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, Jiangxi Province, China
| | - Fangqiang Liu
- Department of Cariology and Endodontics, The Affiliated Stomatological Hospital of Jiujiang University, Jiujiang, Jiangxi Province, China
| | - Hong Li
- Department of Stomatology, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, China
| | - Zhe Liu
- Jiangxi Province Key Laboratory of Oral Biomedicine, Nanchang, Jiangxi Province, China
- Jiangxi Province Clinical Research Center for Oral Diseases, Nanchang, Jiangxi Province, China
- Department of General Dentistry, The Affiliated Stomatological Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, China
| |
Collapse
|
2
|
Bailey L, Stone LD, Gonzalez ML, Thomas CM, Jeyarajan H, Warram JM, Panuganti B. Panitumumab-IRDye800 Improves Laryngeal Tumor Mapping During Transoral Laser Microsurgery. Laryngoscope 2024; 134:1837-1841. [PMID: 37860983 PMCID: PMC10947975 DOI: 10.1002/lary.31078] [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: 07/27/2023] [Revised: 09/12/2023] [Accepted: 09/15/2023] [Indexed: 10/21/2023]
Abstract
Transoral laser microsurgery represents the primary surgical modality for early laryngeal cancers with oncologic outcomes equivalent to radiotherapy. Accurate tumor mapping and margin assessment can be difficult, however, particularly during piecemeal or ablative resections, and for tumors with a wider geographic footprint. Tumor-targeted fluorescence-guided surgery in patients with head and neck cancer has empirically improved tumor and margin identification; this case details, for the first time, a fluorescence-guided surgical resection of a T2N0M0 transglottic tumor using panitumumab-IRDye800, an epidermal growth factor receptor monoclonal antibody covalently linked to near-infrared (NIR) dye. Laryngoscope, 134:1837-1841, 2024.
Collapse
Affiliation(s)
- Luke Bailey
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Logan D Stone
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Manuel L Gonzalez
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Carissa M Thomas
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Hari Jeyarajan
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Jason M Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| | - Bharat Panuganti
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama, USA
| |
Collapse
|
3
|
Wang C, Hodge S, Ravi D, Chen EY, Hoopes PJ, Tichauer KM, Samkoe KS. Rapid and Quantitative Intraoperative Pathology-Assisted Surgery by Paired-Agent Imaging-Derived Confidence Map. Mol Imaging Biol 2023; 25:190-202. [PMID: 36315374 DOI: 10.1007/s11307-022-01780-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/04/2022] [Accepted: 10/05/2022] [Indexed: 02/03/2023]
Abstract
PURPOSE In nonmetastatic head and neck cancer treatment, surgical margin status is the most important prognosticator of recurrence and patient survival. Fresh frozen sectioning (FFS) of tissue margins is the standard of care for intraoperative margin assessment. However, FFS is time intensive, and its accuracy is not consistent among institutes. Mapping the epidermal growth factor receptor (EGFR) using paired-agent imaging (PAI) has the potential to provide more consistent intraoperative margin assessment in a fraction of the time as FFS. PROCEDURES PAI was carried out through IV injection of an anti-epidermal growth factor receptor (EGFR) affibody molecule (ABY-029, eIND 122,681) and an untargeted IRDye680LT carboxylate. Imaging was performed on 4 µm frozen sections from three oral squamous cell carcinoma xenograft mouse models (n = 24, 8 samples per cell line). The diagnostic ability and tumor contrast were compared between binding potential, targeted, and untargeted images. Confidence maps were constructed based on group histogram-derived tumor probability curves. Tumor differentiability and contrast by confidence maps were evaluated. RESULTS PAI outperformed ABY-029 and IRDye 680LT alone, demonstrating the highest individual receiver operating characteristic (ROC) curve area under the curve (PAI AUC: 0.91, 0.90, and 0.79) and contrast-to-noise ratio (PAI CNR: 1, 1.1, and 0.6) for FaDu, Det 562, and A253. PAI confidence maps (PAI CM) maintain high tumor diagnostic ability (PAI CMAUC: 0.91, 0.90, and 0.79) while significantly enhancing tumor contrast (PAI CMCNR: 1.5, 1.3, and 0.8) in FaDu, Det 562, and A253. Additionally, the PAI confidence map allows avascular A253 to be differentiated from a healthy tissue with significantly higher contrast than PAI. Notably, PAI does not require additional staining and therefore significantly reduces the tumor delineation time in a 5 [Formula: see text] 5 mm slice from ~ 35 min to under a minute. CONCLUSION This study demonstrated that PAI improved tumor detection in frozen sections with high diagnostic accuracy and rapid analysis times. The novel PAI confidence map improved the contrast in vascular tumors and differentiability in avascular tumors. With a larger database, the PAI confidence map promises to standardize fluorescence imaging in intraoperative pathology-assisted surgery (IPAS).
Collapse
Affiliation(s)
- Cheng Wang
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Sassan Hodge
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Divya Ravi
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Eunice Y Chen
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - P Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Kenneth M Tichauer
- Biomedical Engineering, Illinois Institute of Technology, Chicago, IL, USA
| | - Kimberley S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA.
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
4
|
Bhandari C, Fakhry J, Eroy M, Song JJ, Samkoe K, Hasan T, Hoyt K, Obaid G. Towards Photodynamic Image-Guided Surgery of Head and Neck Tumors: Photodynamic Priming Improves Delivery and Diagnostic Accuracy of Cetuximab-IRDye800CW. Front Oncol 2022; 12:853660. [PMID: 35837101 PMCID: PMC9273965 DOI: 10.3389/fonc.2022.853660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 05/06/2022] [Indexed: 11/17/2022] Open
Abstract
Fluorescence image-guided surgery (IGS) using antibody conjugates of the fluorophore IRDye800CW have revolutionized the surgical debulking of tumors. Cetuximab, an anti-epidermal growth factor receptor (EGFR) monoclonal antibody, conjugated to IRDye800CW (Cet-IRDye800) is the first molecular targeted antibody probe to be used for IGS in head and neck cancer patients. In addition to surgical debulking, Cetuximab-targeted photodynamic therapy (photoimmunotherapy; PIT) is emerging in the clinic as a powerful modality for head and neck tumor photodestruction. A plethora of other photoactivable agents are also in clinical trials for photodynamic-based therapies of head and neck cancer. Considering the vascular and stromal modulating effects of sub-therapeutic photodynamic therapy, namely photodynamic priming (PDP), this study explores the potential synergy between PDP and IGS for a novel photodynamic image-guided surgery (P-IGS) strategy. To the best of our knowledge, this is the first demonstration that PDP of the tumor microenvironment can augment the tumor delivery of full-length antibodies, namely Cet-IRDye800. In this study, we demonstrate a proof-of-concept that PDP primes orthotopic FaDu human head and neck tumors in mice for P-IGS by increasing the delivery of Cet-IRDye800 by up to 138.6%, by expediting its interstitial accumulation by 10.5-fold, and by increasing its fractional tumor coverage by 49.5% at 1 h following Cet-IRDye800 administration. Importantly, PDP improves the diagnostic accuracy of tumor detection by up to 264.2% with respect to vicinal salivary glands at 1 h. As such, PDP provides a time-to-surgery benefit by reducing the time to plateau 10-fold from 25.7 h to 2.5 h. We therefore propose that a pre-operative PDP regimen can expedite and augment the accuracy of IGS-mediated surgical debulking of head and neck tumors and reduce the time-to-IGS. Furthermore, this P-IGS regimen, can also enable a forward-looking post-operative protocol for the photodestruction of unresectable microscopic disease in the surgical bed. Beyond this scope, the role of PDP in the homogenous delivery of diagnostic, theranostic and therapeutic antibodies in solid tumors is of considerable significance to the wider community.
Collapse
Affiliation(s)
- Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - John Fakhry
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Menitte Eroy
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Jane Junghwa Song
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Kimberley Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover, NH, United States
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, United States
- Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| | - Girgis Obaid
- Department of Bioengineering, University of Texas at Dallas, Richardson, TX, United States
| |
Collapse
|
5
|
Guirguis M, Bhandari C, Li J, Eroy M, Prajapati S, Margolis R, Shrivastava N, Hoyt K, Hasan T, Obaid G. Membrane composition is a functional determinant of NIR-activable liposomes in orthotopic head and neck cancer. NANOPHOTONICS 2021; 10:3169-3185. [PMID: 35433177 PMCID: PMC9012185 DOI: 10.1515/nanoph-2021-0191] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Near-infrared (NIR)-activable liposomes containing photosensitizer (PS)-lipid conjugates are emerging as tunable, high-payload, and tumor-selective platforms for photodynamic therapy (PDT)-based theranostics. To date, the impact that the membrane composition of a NIR-activable liposome (the chemical nature and subsequent conformation of PS-lipid conjugates) has on their in vitro and in vivo functionality has not been fully investigated. While their chemical nature is critical, the resultant physical conformation dictates their interactions with the immediate biological environments. Here, we evaluate NIR-activable liposomes containing lipid conjugates of the clinically-used PSs benzoporphyrin derivative (BPD; hydrophobic, membrane-inserting conformation) or IRDye 700DX (hydrophilic, membrane-protruding conformation) and demonstrate that membrane composition is critical for their function as tumor-selective PDT-based platforms. The PS-lipid conformations were primarily dictated by the varying solubilities of the two PSs and assisted by their lipid conjugation sites. Conformation was further validated by photophysical analysis and computational predictions of PS membrane partitioning (topological polar surface area [tPSA], calculated octanol/water partition [cLogP], and apparent biomembrane permeability coefficient [Papp]). Results show that the membrane-protruding lipo-IRDye700DX exhibits 5-fold more efficient photodynamic generation of reactive molecular species (RMS), 12-fold expedited phototriggered burst release of entrap-ped agents, and 15-fold brighter fluorescence intensity as compared to the membrane-inserting lipo-BPD-PC (phosphatidylcholine conjugate). Although the membrane-inserting lipo-BPD-PC exhibits less efficient photo-dynamic generation of RMS, it allows for more sustained phototriggered release, 10-fold greater FaDu cancer cell phototoxicity, and 7.16-fold higher tumor-selective delivery in orthotopic mouse FaDu head and neck tumors. These critical insights pave the path for the rational design of emerging NIR-activable liposomes, whereby functional consequences of membrane composition can be tailored toward a specific therapeutic purpose.
Collapse
Affiliation(s)
- Mina Guirguis
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Chanda Bhandari
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Junjie Li
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Menitte Eroy
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Sushant Prajapati
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Ryan Margolis
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Navadeep Shrivastava
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Kenneth Hoyt
- Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA
| | - Tayyaba Hasan
- Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston 02114, Massachusetts, USA; and Division of Health Sciences and Technology, Harvard University and Massachusetts Institute of Technology, Cambridge 02139, Massachusetts, USA
| | - Girgis Obaid
- Corresponding author: Girgis Obaid, Department of Bioengineering, University of Texas at Dallas, Richardson 75080, Texas, USA,
| |
Collapse
|
6
|
van Schaik JE, Halmos GB, Witjes MJH, Plaat BEC. An overview of the current clinical status of optical imaging in head and neck cancer with a focus on Narrow Band imaging and fluorescence optical imaging. Oral Oncol 2021; 121:105504. [PMID: 34454339 DOI: 10.1016/j.oraloncology.2021.105504] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Revised: 07/25/2021] [Accepted: 08/18/2021] [Indexed: 11/28/2022]
Abstract
Early and accurate identification of head and neck squamous cell carcinoma (HNSCC) is important to improve treatment outcomes and prognosis. New optical imaging techniques may assist in both the diagnostic process as well as in the operative setting by real-time visualization and delineation of tumor. Narrow Band Imaging (NBI) is an endoscopic technique that uses blue and green light to enhance mucosal and submucosal blood vessels, leading to better detection of (pre)malignant lesions showing aberrant blood vessel patterns. Fluorescence optical imaging makes use of near-infrared fluorescent agents to visualize and delineate HNSCC, resulting in fewer positive surgical margins. Targeted fluorescent agents, such as fluorophores conjugated to antibodies, show the most promising results. The aim of this review is: (1) to provide the clinical head and neck surgeon an overview of the current clinical status of various optical imaging techniques in head and neck cancer; (2) to provide an in-depth review of NBI and fluorescence optical imaging, as these techniques have the highest potential for clinical implementation; and (3) to describe future improvements and developments within the field of these two techniques.
Collapse
Affiliation(s)
- Jeroen E van Schaik
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands.
| | - Gyorgy B Halmos
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Max J H Witjes
- Department of Oral and Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, the Netherlands
| | - Boudewijn E C Plaat
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| |
Collapse
|
7
|
Cohen-Kaplan V, Ilan N, Vlodavsky I. Heparanase Loosens E-Cadherin-Mediated Cell-Cell Contact via Activation of Src. Front Oncol 2020; 10:2. [PMID: 32038981 PMCID: PMC6990126 DOI: 10.3389/fonc.2020.00002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 01/02/2020] [Indexed: 01/18/2023] Open
Abstract
Activity of heparanase, responsible for cleavage of heparan sulfate (HS), is strongly implicated in tumor metastasis. This is due primarily to remodeling of the extracellular matrix (ECM) that becomes more prone to invasion by metastatic tumor cells. In addition, heparanase promotes the development of blood and lymph vessels that mobilize disseminated cells to distant organs. Here, we provide evidence for an additional mechanism by which heparanase affects cell motility, namely the destruction of E-cadherin based adherent junctions (AJ). We found that overexpression of heparanase or its exogenous addition results in reduced E-cadherin levels in the cell membrane. This was associated with a substantial increase in the phosphorylation levels of E-cadherin, β-catenin, and p120-catenin, the latter recognized as a substrate of Src. Indeed, we found that Src phosphorylation is increased in heparanase overexpressing cells, associating with a marked decrease in the interaction of E-cadherin with β-catenin, which is instrumental for AJ integrity and cell-cell adhesion. Notably, the association of E-cadherin with β-catenin in heparanase overexpressing cells was restored by Src inhibitor, along with reduced cell migration. These results imply that heparanase promotes tumor metastasis by virtue of its enzymatic activity responsible for remodeling of the ECM, and by signaling aspects that result in Src-mediated phosphorylation of E-cadherin/catenins and loosening of cell-cell contacts that are required for maintaining the integrity of epithelial sheets.
Collapse
Affiliation(s)
- Victoria Cohen-Kaplan
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center (TICC), Technion, Haifa, Israel
| | - Neta Ilan
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center (TICC), Technion, Haifa, Israel
| | - Israel Vlodavsky
- Rappaport Faculty of Medicine, Technion Integrated Cancer Center (TICC), Technion, Haifa, Israel
| |
Collapse
|
8
|
Electric Field Based Dressing Disrupts Mixed-Species Bacterial Biofilm Infection and Restores Functional Wound Healing. Ann Surg 2020; 269:756-766. [PMID: 29099398 DOI: 10.1097/sla.0000000000002504] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVE This study was designed to employ electroceutical principles, as an alternative to pharmacological intervention, to manage wound biofilm infection. Mechanism of action of a United States Food and Drug Administration-cleared wireless electroceutical dressing (WED) was tested in an established porcine chronic wound polymicrobial biofilm infection model involving inoculation with Pseudomonas aeruginosa PAO1 and Acinetobacter baumannii 19606. BACKGROUND Bacterial biofilms represent a major wound complication. Resistance of biofilm toward pharmacologic interventions calls for alternative therapeutic strategies. Weak electric field has anti-biofilm properties. We have previously reported the development of WED involving patterned deposition of Ag and Zn on fabric. When moistened, WED generates a weak electric field without any external power supply and can be used as any other disposable dressing. METHODS WED dressing was applied within 2 hours of wound infection to test its ability to prevent biofilm formation. Alternatively, WED was applied after 7 days of infection to study disruption of established biofilm. Wounds were treated with placebo dressing or WED twice a week for 56 days. RESULTS Scanning electron microscopy demonstrated that WED prevented and disrupted wound biofilm aggregates. WED accelerated functional wound closure by restoring skin barrier function. WED blunted biofilm-induced expression of (1) P. aeruginosa quorum sensing mvfR (pqsR), rhlR and lasR genes, and (2) miR-9 and silencing of E-cadherin. E-cadherin is critically required for skin barrier function. Furthermore, WED rescued against biofilm-induced persistent inflammation by circumventing nuclear factor kappa B activation and its downstream cytokine responses. CONCLUSION This is the first pre-clinical porcine mechanistic study to recognize the potential of electroceuticals as an effective platform technology to combat wound biofilm infection.
Collapse
|
9
|
Ding H, Kothandaraman S, Gong L, Wright CL, Pan Q, Teknos T, Tweedle MF. Novel Peptide NIRF Optical Surgical Navigation Agents for HNSCC. MOLECULES (BASEL, SWITZERLAND) 2019; 24:molecules24173070. [PMID: 31450798 PMCID: PMC6749330 DOI: 10.3390/molecules24173070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/22/2019] [Revised: 08/19/2019] [Accepted: 08/20/2019] [Indexed: 12/24/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) survival rates have not improved in a decade, with a 63% 5-year recurrence rate after surgery, making HNSCC a compelling indication for optical surgical navigation (OSN). A promising peptide, HN1, targeted and internalized in human HNSCC cells in multiple laboratories, but was slow (24 h) to accumulate. We modified HN1 and explored structural variables to improve the uptake kinetics and create IRdye800 adducts useful for OSN. Eleven new molecules were synthesized and characterized chemically, in human HNSCC cells (Cal 27), and in HNSCC xenograft mice. Cal 27 flank xenografts in Balb/c nude mice were imaged for 3-48 h after 40 nmol intravenous doses of IR800-labeled molecules. Cell uptake kinetics in the 1-2 h window incubated at 1-10 μM were independent of the dye label (FITC, Cy5, or IR800), but increased markedly with additional N-terminal lipophilic substitution, and after resequencing the peptide to separate polar amino acids and move the lysine-dye more centrally. Microscopy confirmed the strong Cal 27 cell binding and demonstrated primarily cytosolic and membrane localization of the fastest peptide, 4Iphf-HN17. 4Iph-HN17-IR800 showed 26-fold greater rate of uptake in cells than HN1-IR800, and far stronger OSN imaging intensity and tumor to background contrast in mice, suggesting that the new peptide is a promising candidate for OSN of HNSCC.
Collapse
Affiliation(s)
- Haiming Ding
- Department of Radiology, The Wright Center of Innovation in Biomedical Imaging, The Ohio State University, Columbus, OH 43210, USA
| | - Shankaran Kothandaraman
- Department of Radiology, The Wright Center of Innovation in Biomedical Imaging, The Ohio State University, Columbus, OH 43210, USA
| | - Li Gong
- Department of Radiology, The Wright Center of Innovation in Biomedical Imaging, The Ohio State University, Columbus, OH 43210, USA
| | - Chadwick L Wright
- Department of Radiology, The Wright Center of Innovation in Biomedical Imaging, The Ohio State University, Columbus, OH 43210, USA
| | - Quintin Pan
- Seidman Cancer Center, University Hospitals, Cleveland, OH 44106, USA
| | - Theodore Teknos
- Seidman Cancer Center, University Hospitals, Cleveland, OH 44106, USA
| | - Michael F Tweedle
- Department of Radiology, The Wright Center of Innovation in Biomedical Imaging, The Ohio State University, Columbus, OH 43210, USA.
| |
Collapse
|
10
|
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: 2.4] [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
|
11
|
Feroldi F, Verlaan M, Knaus H, Davidoiu V, Vugts DJ, van Dongen GAMS, Molthoff CFM, de Boer JF. High resolution combined molecular and structural optical imaging of colorectal cancer in a xenograft mouse model. BIOMEDICAL OPTICS EXPRESS 2018; 9:6186-6204. [PMID: 31065422 PMCID: PMC6491025 DOI: 10.1364/boe.9.006186] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Revised: 11/01/2018] [Accepted: 11/06/2018] [Indexed: 05/08/2023]
Abstract
With the emergence of immunotherapies for cancer treatment, there is a rising clinical need to visualize the tumor microenvironment (TME) non-invasively in detail, which could be crucial to predict the efficacy of therapy. Nuclear imaging techniques enable whole-body imaging but lack the required spatial resolution. Conversely, near-infrared immunofluorescence (immuno-NIRF) is able to reveal tumor cells and/or other cell subsets in the TME by targeting the expression of a specific membrane receptor with fluorescently labeled monoclonal antibodies (mAb). Optical coherence tomography (OCT) provides three-dimensional morphological imaging of tissues without exogenous contrast agents. The combination of the two allows molecular and structural contrast at a resolution of ~15 µm, allowing for the specific location of a cell-type target with immuno-NIRF as well as revealing the three-dimensional architectural context with OCT. For the first time, combined immuno-NIRF and OCT of a tumor is demonstrated in situ in a xenograft mouse model of human colorectal cancer, targeted by a clinically-safe fluorescent mAb, revealing unprecedented details of the TME. A handheld scanner for ex vivo examination and an endoscope designed for imaging bronchioles in vivo are presented. This technique promises to complement nuclear imaging for diagnosing cancer invasiveness, precisely determining tumor margins, and studying the biodistribution of newly developed antibodies in high detail.
Collapse
Affiliation(s)
- Fabio Feroldi
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| | - Mariska Verlaan
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Helene Knaus
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| | - Valentina Davidoiu
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| | - Danielle J. Vugts
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Guus A. M. S. van Dongen
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Carla F. M. Molthoff
- Amsterdam UMC, Vrije Universiteit Amsterdam, Department of Radiology & Nuclear Medicine, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
| | - Johannes F. de Boer
- Department of Physics and Astronomy, LaserLaB Amsterdam, VU University Amsterdam, de Boelelaan 1081, 1081HV, Amsterdam, The Netherlands
| |
Collapse
|
12
|
Lwin TM, Hoffman RM, Bouvet M. Advantages of patient-derived orthotopic mouse models and genetic reporters for developing fluorescence-guided surgery. J Surg Oncol 2018; 118:253-264. [PMID: 30080930 PMCID: PMC6146062 DOI: 10.1002/jso.25150] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2018] [Accepted: 06/04/2018] [Indexed: 12/16/2022]
Abstract
Fluorescence-guided surgery can enhance the surgeon's ability to achieve a complete oncologic resection. There are a number of tumor-specific probes being developed with many preclinical mouse models to evaluate their efficacy. The current review discusses the different preclinical mouse models in the setting of probe evaluation and highlights the advantages of patient-derived orthotopic xenografts (PDOX) mouse models and genetic reporters to develop fluorescence-guided surgery.
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
- Department of Surgery, VA Medical Center, San Diego, CA
| |
Collapse
|
13
|
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: 3.3] [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
|
14
|
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: 27] [Impact Index Per Article: 4.5] [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
|
15
|
Samkoe KS, Gunn JR, Marra K, Hull SM, Moodie KL, Feldwisch J, Strong TV, Draney DR, Hoopes PJ, Roberts DW, Paulsen K, Pogue BW. Toxicity and Pharmacokinetic Profile for Single-Dose Injection of ABY-029: a Fluorescent Anti-EGFR Synthetic Affibody Molecule for Human Use. Mol Imaging Biol 2018; 19:512-521. [PMID: 27909986 DOI: 10.1007/s11307-016-1033-y] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
PURPOSE ABY-029, a synthetic Affibody peptide, Z03115-Cys, labeled with a near-infrared fluorophore, IRDye® 800CW, targeting epidermal growth factor receptor (EGFR) has been produced under good manufacturing practices for a US Food and Drug Administration-approved first-in-use human study during surgical resection of glioma, as well as other tumors. Here, the pharmacology, phototoxicity, receptor activity, and biodistribution studies of ABY-029 were completed in rats, prior to the intended human use. PROCEDURES Male and female Sprague Dawley rats were administered a single intravenous dose of varying concentrations (0, 245, 2449, and 24,490 μg/kg corresponding to 10×, 100×, and 1000× an equivalent human microdose level) of ABY-029 and observed for up to 14 days. Histopathological assessment of organs and tissues, clinical chemistry, and hematology were performed. In addition, pharmacokinetic clearance and biodistribution of ABY-029 were studied in subgroups of the animals. Phototoxicity and ABY-029 binding to human and rat EGFR were assessed in cell culture and on immobilized receptors, respectively. RESULTS Histopathological assessment and hematological and clinical chemistry analysis demonstrated that single-dose ABY-029 produced no pathological evidence of toxicity at any dose level. No phototoxicity was observed in EGFR-positive and EGFR-negative glioma cell lines. Binding strength and pharmacokinetics of the anti-EGFR Affibody molecules were retained after labeling with the dye. CONCLUSION Based on the successful safety profile of ABY-029, the 1000× human microdose 24.5 mg/kg was identified as the no observed adverse effect level following intravenous administration. Conserved binding strength and no observed light toxicity also demonstrated ABY-029 safety for human use.
Collapse
Affiliation(s)
- Kimberley S Samkoe
- Department of Surgery, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, 03755, USA. .,Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA.
| | - Jason R Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Kayla Marra
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Sally M Hull
- Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Karen L Moodie
- Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH, 03755, USA
| | | | - Theresa V Strong
- Department of Medicine, University of Alabama at Birmingham School of Medicine, Birmingham, AL, 35294, USA
| | | | - P Jack Hoopes
- Department of Surgery, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, 03755, USA.,Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA.,Center for Comparative Medicine and Research, Dartmouth College, Hanover, NH, 03755, USA
| | - David W Roberts
- Department of Surgery, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, 03755, USA.,Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Keith Paulsen
- Department of Surgery, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, 03755, USA.,Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA
| | - Brian W Pogue
- Department of Surgery, Geisel School of Medicine at Dartmouth, Dartmouth College, Hanover, NH, 03755, USA. .,Thayer School of Engineering, Dartmouth College, Hanover, NH, 03755, USA.
| |
Collapse
|
16
|
Mizushima T, Ohnishi S, Shimizu Y, Hatanaka Y, Hatanaka KC, Kuriki Y, Kamiya M, Homma A, Yamamoto K, Ono S, Urano Y, Sakamoto N. Rapid detection of superficial head and neck squamous cell carcinoma by topically spraying fluorescent probe targeting dipeptidyl peptidase-IV. Head Neck 2018; 40:1466-1475. [PMID: 29509281 DOI: 10.1002/hed.25126] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 11/15/2017] [Accepted: 01/26/2018] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND A fluorescent probe glutamylprolyl hydroxymethyl rhodamine green (EP-HMRG), which becomes fluorescent after cleavage by dipeptidyl peptidase-IV (DPP-IV), has been reported to be useful for the detection of esophageal cancer. Thus, we investigated whether head and neck squamous cell carcinoma (HNSCC) can be detected by spraying EP-HMRG. METHODS Fluorescence imaging of 17 cases of HNSCCs resected using endoscopic or surgical resection was performed ex vivo after spraying EP-HMRG, and then the fluorescence intensity of the tumors and normal mucosa were measured. RESULTS Iodine-voiding lesions became fluorescent within a few minutes after the application of EP-HMRG in 12 resected tumors without a history of radiotherapy but this was not observed in the normal mucosa. Fluorescence intensity in tumor lesions was significantly higher than normal lesions. However, 5 other tumors that developed after radiotherapy did not have sufficient contrast against normal mucosa. CONCLUSION Fluorescence imaging with EP-HMRG would be useful for rapid detection of superficial HNSCC without a history of radiotherapy.
Collapse
Affiliation(s)
- Takeshi Mizushima
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shunsuke Ohnishi
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Yuichi Shimizu
- Division of Endoscopy, Hokkaido University Hospital, Sapporo, Japan
| | - Yutaka Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Kanako C Hatanaka
- Department of Surgical Pathology, Hokkaido University Hospital, Sapporo, Japan
| | - Yugo Kuriki
- Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Mako Kamiya
- Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan
| | - Akihiro Homma
- Department of Otolaryngology - Head and Neck Surgery, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Keiko Yamamoto
- Division of Endoscopy, Hokkaido University Hospital, Sapporo, Japan
| | - Shouko Ono
- Division of Endoscopy, Hokkaido University Hospital, Sapporo, Japan
| | - Yasuteru Urano
- Laboratory of Chemical Biology and Molecular Imaging, Graduate School of Medicine, The University of Tokyo, Tokyo, Japan.,Japan Agency for Medical Research and Development (AMED)-CREST, Tokyo, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
17
|
Samkoe KS, Bates BD, Elliott JT, LaRochelle E, Gunn JR, Marra K, Feldwisch J, Ramkumar DB, Bauer DF, Paulsen KD, Pogue BW, Henderson ER. Application of Fluorescence-Guided Surgery to Subsurface Cancers Requiring Wide Local Excision: Literature Review and Novel Developments Toward Indirect Visualization. Cancer Control 2018; 25:1073274817752332. [PMID: 29334791 PMCID: PMC5933571 DOI: 10.1177/1073274817752332] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2016] [Accepted: 12/05/2017] [Indexed: 01/08/2023] Open
Abstract
The excision of tumors by wide local excision is challenging because the mass must be removed entirely without ever viewing it directly. Positive margin rates in sarcoma resection remain in the range of 20% to 35% and are associated with increased recurrence and decreased survival. Fluorescence-guided surgery (FGS) may improve surgical accuracy and has been utilized in other surgical specialties. ABY-029, an anti-epidermal growth factor receptor Affibody molecule covalently bound to the near-infrared fluorophore IRDye 800CW, is an excellent candidate for future FGS applications in sarcoma resection; however, conventional methods with direct surface tumor visualization are not immediately applicable. A novel technique involving imaging through a margin of normal tissue is needed. We review the past and present applications of FGS and present a novel concept of indirect FGS for visualizing tumor through a margin of normal tissue and aiding in excising the entire lesion as a single, complete mass with tumor-free margins.
Collapse
Affiliation(s)
- Kimberley S. Samkoe
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Brent D. Bates
- Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | | | - Ethan LaRochelle
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Jason R. Gunn
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Kayla Marra
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | | | - Dipak B. Ramkumar
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - David F. Bauer
- Department of Surgery, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
| | - Keith D. Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Brian W. Pogue
- Thayer School of Engineering, Dartmouth College, Hanover, NH, USA
| | - Eric R. Henderson
- Department of Orthopaedics, Dartmouth-Hitchcock Medical Center, Lebanon, NH, USA
- White River Junction VA Medical Center, White River Junction, VT, USA
| |
Collapse
|
18
|
Moore LS, Rosenthal EL, Chung TK, de Boer E, Patel N, Prince AC, Korb ML, Walsh EM, Young ES, Stevens TM, Withrow KP, Morlandt AB, Richman JS, Carroll WR, Zinn KR, Warram JM. Characterizing the Utility and Limitations of Repurposing an Open-Field Optical Imaging Device for Fluorescence-Guided Surgery in Head and Neck Cancer Patients. J Nucl Med 2017; 58:246-251. [PMID: 27587708 PMCID: PMC5288741 DOI: 10.2967/jnumed.115.171413] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2015] [Accepted: 08/10/2016] [Indexed: 01/24/2023] Open
Abstract
The purpose of this study was to assess the potential of U.S. Food and Drug Administration-cleared devices designed for indocyanine green-based perfusion imaging to identify cancer-specific bioconjugates with overlapping excitation and emission wavelengths. Recent clinical trials have demonstrated potential for fluorescence-guided surgery, but the time and cost of the approval process may impede clinical translation. To expedite this translation, we explored the feasibility of repurposing existing optical imaging devices for fluorescence-guided surgery. METHODS Consenting patients (n = 15) scheduled for curative resection were enrolled in a clinical trial evaluating the safety and specificity of cetuximab-IRDye800 (NCT01987375). Open-field fluorescence imaging was performed preoperatively and during the surgical resection. Fluorescence intensity was quantified using integrated instrument software, and the tumor-to-background ratio characterized fluorescence contrast. RESULTS In the preoperative clinic, the open-field device demonstrated potential to guide preoperative mapping of tumor borders, optimize the day of surgery, and identify occult lesions. Intraoperatively, the device demonstrated robust potential to guide surgical resections, as all peak tumor-to-background ratios were greater than 2 (range, 2.2-14.1). Postresection wound bed fluorescence was significantly less than preresection tumor fluorescence (P < 0.001). The repurposed device also successfully identified positive margins. CONCLUSION The open-field imaging device was successfully repurposed to distinguish cancer from normal tissue in the preoperative clinic and throughout surgical resection. This study illuminated the potential for existing open-field optical imaging devices with overlapping excitation and emission spectra to be used for fluorescence-guided surgery.
Collapse
Affiliation(s)
- Lindsay S Moore
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, California
| | - Thomas K Chung
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Esther de Boer
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
- Department of Surgery, University of Groningen, Groningen, The Netherlands
| | - Neel Patel
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Andrew C Prince
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Melissa L Korb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Erika M Walsh
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - E Scott Young
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Todd M Stevens
- Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kirk P Withrow
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Anthony B Morlandt
- Department of Oral and Maxillofacial Surgery, University of Alabama at Birmingham, Birmingham, Alabama; and
| | - Joshua S Richman
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - William R Carroll
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kurt R Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jason M Warram
- Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
19
|
Sweeny L, Prince A, Patel N, Moore LS, Rosenthal EL, Hughley BB, Warram JM. Antiangiogenic antibody improves melanoma detection by fluorescently labeled therapeutic antibodies. Laryngoscope 2016; 126:E387-E395. [PMID: 27576611 DOI: 10.1002/lary.26215] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2016] [Revised: 06/14/2016] [Accepted: 07/07/2016] [Indexed: 12/19/2022]
Abstract
OBJECTIVE Evaluate if vascular normalization with an antiangiogenic monoclonal antibody improves detection of melanoma using fluorescently labeled antibody-based imaging. STUDY DESIGN Preclinical. METHODS Panitumumab and control IgG were covalently linked to a near-infrared fluorescent probe (IRDye800CW). Immunodeficient mice with ear xenografts of melanoma cell lines (A375 and SKMEL5) were systemically injected (200 μg, tail vein) with either IgG-IRDye800CW, panitumumab-IRDye800CW, or a combination (bevacizumab [5mg/kg], administered 72 hours prepanitumumab-IRDye800CW) (n = 5). Primary tumors were imaged with open-field (LUNA, Novadaq, Toronto, Ontario, Canada) and closed-field (Pearl, LI-COR Biosciences, Lincoln, NB) imaging devices. Postresection, the concentration of labeled antibody within the tumor (μg/g) was calculated using normalized standards. RESULTS The mean fluorescence within the melanoma tumors was greater for the combination group compared to panitumumab alone for both cell lines (P < 0.001). The tumor-to-background ratio (TBR) for the A375 tumors was greater for the combination (3.4-7.1) compared to the panitumumab alone (3.2-5.0) (P = 0.04). The TBR for SKMEL5 tumors was greater for the combination (2.4-6.0) compared to the panitumumab alone (2.2-3.9) (P = 0.02). Within A375 tumors, the concentration was lower for panitumumab (0.51 μg/g) compared to combination group (0.68 μg/g) (P = 0.036). Within SKMEL5 tumors, the concentration was lower for panitumumab (0.0.17 μg/g) compared to combination group (0.35 μg/g) (P = 0.048). Residual tumor (1.0-0.2 mg) could be differentiated from background in both panitumumab and combination groups. For both cell lines, panitumumab and combination groups had greater mean fluorescence of the tumor compared to control IgG. CONCLUSION The addition of antiangiogenic therapy improves uptake of fluorescently labeled monoclonal antibodies within melanoma tumors. Clinical translation could improve detection of melanoma intraoperatively, reducing positive margins and sparing normal tissue. LEVEL OF EVIDENCE NA Laryngoscope, 126:E387-E395, 2016.
Collapse
Affiliation(s)
- Larissa Sweeny
- Department of Otolaryngology-Head and Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Andrew Prince
- the University of Alabama School of Medicine at Birmingham, Birmingham, Alabama, U.S.A
| | - Neel Patel
- the Department of Psychiatry, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Lindsay S Moore
- Department of Otolaryngology-Head and Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, California, U.S.A
| | - Brian B Hughley
- Department of Otolaryngology-Head and Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| | - Jason M Warram
- Department of Otolaryngology-Head and Neck Surgery, University of Alabama at Birmingham, Birmingham, Alabama, U.S.A
| |
Collapse
|
20
|
Bekeny JR, Ozer E. Transoral robotic surgery frontiers. World J Otorhinolaryngol Head Neck Surg 2016; 2:130-135. [PMID: 29204557 PMCID: PMC5698526 DOI: 10.1016/j.wjorl.2016.05.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2016] [Accepted: 05/12/2016] [Indexed: 10/30/2022] Open
Abstract
Transoral robotic surgery is a exciting field that continues to develop and push the boundaries of current procedural ability and challenges historical treatment paradigms. With the first use of a surgical robot in 1985, to the first clinical use of the robot transorally in 2005, there was some lag in adoption of robotic techniques in the head and neck region. However, since 2005 transoral robotic surgery has rapidly gained momentum amongst head and neck surgeons. With FDA approval of the da Vinci robot in 2009, transoral robotic surgery is currently offered as a treatment modality for malignant and nonmalignant disease of the head and neck region. This new technology is being used to reconsider historical treatment paradigms for malignancies of the upper aerodigestive tract due to the fact that minimally invasive surgical access to the oropharynx and larynx has been improved. Along with this enhanced access have come innovative procedures and uses of the technology for multiple facets of head and neck disease. Technology continues to improve and innovation in surgical robotics is expected to continue as more companies attempt to capture this market. This article aims to provide a view at the landscape of transoral robotic surgery and explore the future frontiers.
Collapse
Affiliation(s)
- James R Bekeny
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Medical Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| | - Enver Ozer
- Department of Otolaryngology-Head and Neck Surgery, The Ohio State University Medical Center, The James Cancer Hospital and Solove Research Institute, Columbus, OH 43210, USA
| |
Collapse
|
21
|
Assessment of indocyanine green–labeled cetuximab to detect xenografted head and neck cancer cell lines. Otolaryngol Head Neck Surg 2016; 137:729-34. [DOI: 10.1016/j.otohns.2007.06.736] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2006] [Revised: 04/17/2007] [Accepted: 06/26/2007] [Indexed: 11/18/2022]
Abstract
Objective The aim of this study is to determine the efficacy of indocyanine green (ICG) conjugated to antiepidermal growth factor receptor antibody (cetuximab) to image head and neck cancer. Study Design Mice (n = 3) were injected with unconjugated ICG and imaged at 100-second intervals for a total of 1000 seconds to assess imaging characteristics. Mice (n = 10) xenografted with SCC-1 cells were then systemically injected with cetuximab conjugated to indocyanine green and imaged over a 72-hour period. To assess the sensitivity and specificity, xenografted tumors underwent subtotal resections and then were assessed for residual disease by fluorescence stereomicroscopy and confirmed by histology. Results Tumors demonstrated excellent fluorescence 24 hours after injection of cetuximab-ICG. There was a direct relationship between fluorescence and the given dose of cetuximab-ICG. Following subtotal resection, we found fluorescence correlated with a sensitivity of 78.4% and specificity of 96%. Conclusions This study provides evidence that supports further preclinical investigation of cetuximab in the evaluation of surgical margins, but linkage to ICG lacks the sensitivity for use in a clinical setting.
Collapse
|
22
|
Hussain T, Savariar EN, Diaz-Perez JA, Messer K, Pu M, Tsien RY, Nguyen QT. Surgical molecular navigation with ratiometric activatable cell penetrating peptide for intraoperative identification and resection of small salivary gland cancers. Head Neck 2016; 38:715-23. [PMID: 25521629 PMCID: PMC4472578 DOI: 10.1002/hed.23946] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2014] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND We evaluated the use of intraoperative fluorescence guidance by enzymatically cleavable ratiometric activatable cell-penetrating peptide (RACPPPLGC(Me)AG) containing Cy5 as a fluorescent donor and Cy7 as a fluorescent acceptor for salivary gland cancer surgery in a mouse model. METHODS Surgical resection of small parotid gland cancers in mice was performed with fluorescence guidance or white light (WL) imaging alone. Tumor identification accuracy, operating time, and tumor-free survival were compared. RESULTS RACPP guidance aided tumor detection (positive histology in 90% [27/30] vs 48% [15/31] for WL; p < .001). An approximate 25% ratiometric signal increase as the threshold to distinguish between tumor and adjacent tissue, yielded >90% detection sensitivity and specificity. Operating time was reduced by 54% (p < .001), and tumor-free survival was increased with RACPP guidance (p = .025). CONCLUSION RACPP provides real-time intraoperative guidance leading to improved survival. Ratiometric signal thresholds can be set according to desired detection accuracy levels for future RACPP applications.
Collapse
Affiliation(s)
- Timon Hussain
- Division of Head and Neck Surgery, University of California San Diego
| | | | | | - Karen Messer
- Division of Biostatistics, Moores Cancer Center, University of California San Diego
| | - Minya Pu
- Division of Biostatistics, Moores Cancer Center, University of California San Diego
| | - Roger Y. Tsien
- Department of Pharmacology, University of California San Diego
- Howard Hughes Medical Institute, University of California San Diego
| | - Quyen T. Nguyen
- Division of Head and Neck Surgery, University of California San Diego
| |
Collapse
|
23
|
Rosenthal EL, Warram JM, de Boer E, Basilion JP, Biel MA, Bogyo M, Bouvet M, Brigman BE, Colson YL, DeMeester SR, Gurtner GC, Ishizawa T, Jacobs PM, Keereweer S, Liao JC, Nguyen QT, Olson JM, Paulsen KD, Rieves D, Sumer BD, Tweedle MF, Vahrmeijer AL, Weichert JP, Wilson BC, Zenn MR, Zinn KR, van Dam GM. Successful Translation of Fluorescence Navigation During Oncologic Surgery: A Consensus Report. J Nucl Med 2015; 57:144-50. [PMID: 26449839 DOI: 10.2967/jnumed.115.158915] [Citation(s) in RCA: 106] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2015] [Accepted: 09/24/2015] [Indexed: 11/16/2022] Open
Abstract
Navigation with fluorescence guidance has emerged in the last decade as a promising strategy to improve the efficacy of oncologic surgery. To achieve routine clinical use, the onus is on the surgical community to objectively assess the value of this technique. This assessment may facilitate both Food and Drug Administration approval of new optical imaging agents and reimbursement for the imaging procedures. It is critical to characterize fluorescence-guided procedural benefits over existing practices and to elucidate both the costs and the safety risks. This report is the result of a meeting of the International Society of Image Guided Surgery (www.isigs.org) on February 6, 2015, in Miami, Florida, and reflects a consensus of the participants' opinions. Our objective was to critically evaluate the imaging platform technology and optical imaging agents and to make recommendations for successful clinical trial development of this highly promising approach in oncologic surgery.
Collapse
Affiliation(s)
- Eben L Rosenthal
- Department of Otolaryngology, Stanford University, Stanford, California
| | - Jason M Warram
- Division of Otolaryngology, Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Esther de Boer
- Division of Otolaryngology, Department of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - James P Basilion
- Case Center for Imaging Research, NFCR Center for Molecular Imaging, Department of Radiology Case Western Reserve University, Cleveland, Ohio
| | - Merrill A Biel
- Department of Otolaryngology-Head and Neck Surgery, University of Minnesota, Minneapolis, Minnesota
| | - Matthew Bogyo
- Department of Pathology, Stanford School of Medicine, Stanford, California Department of Cancer Biology Program, Stanford School of Medicine, Stanford, California
| | - Michael Bouvet
- Department of Surgery, University of California San Diego, La Jolla, California
| | - Brian E Brigman
- Division of Orthopaedic Surgery, Department of Surgery, Duke University Medical Center, Durham, North Carolina
| | - Yolonda L Colson
- Division of Thoracic Surgery, Brigham and Women's Hospital, Boston, Massachusetts
| | - Steven R DeMeester
- Department of Surgery, Keck School of Medicine of the University of Southern California, Los Angeles, California
| | - Geoffrey C Gurtner
- Hagey Laboratory for Pediatric Regenerative Medicine, Division of Plastic and Reconstructive Surgery, Department of Surgery, Stanford University School of Medicine, Stanford, California
| | - Takeaki Ishizawa
- Artificial Organ and Transplantation Surgery Division, Department of Surgery, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | | | - Stijn Keereweer
- Department of Otorhinolaryngology Head and Neck Surgery, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Joseph C Liao
- Department of Urology and Bio-X Program, Stanford University School of Medicine, Stanford, California
| | - Quyen T Nguyen
- Division of Head and Neck Surgery, University of California San Diego, San Diego, California
| | - James M Olson
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington Seattle Children's Hospital & Regional Medical Center, Seattle, Washington
| | - Keith D Paulsen
- Department of Diagnostic Radiology, Geisel School of Medicine, Dartmouth College, Hanover, New Hampshire
| | | | - Baran D Sumer
- Department of Otolaryngology, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael F Tweedle
- Department of Radiology, The Wright Center for Innovation in Biomedical Imaging, The Ohio State University, Columbus, Ohio
| | | | - Jamey P Weichert
- Department of Radiology, University of Wisconsin, Madison, Wisconsin
| | - Brian C Wilson
- Princess Margaret Cancer Centre, UHN, Toronto, Ontario, Canada Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Michael R Zenn
- Plastic and Reconstructive Surgery, Duke University Medical Center, Durham, North Carolina; and
| | - Kurt R Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gooitzen M van Dam
- Department of Surgery, Nuclear Medicine and Molecular Imaging and Intensive Care, University Medical Center Groningen, University of Groningen, The Netherlands, on behalf of the International Society of Image Guided Surgery (ISIGS)
| |
Collapse
|
24
|
Wang ZQ, Liu K, Huo ZJ, Li XC, Wang M, Liu P, Pang B, Wang SJ. A cell-targeted chemotherapeutic nanomedicine strategy for oral squamous cell carcinoma therapy. J Nanobiotechnology 2015; 13:63. [PMID: 26427800 PMCID: PMC4591064 DOI: 10.1186/s12951-015-0116-2] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2015] [Accepted: 08/20/2015] [Indexed: 02/01/2023] Open
Abstract
Background Oral squamous cell carcinoma (OSCC) or cancers of oral cavity is one of the most common cancers worldwide with high rate of mortality and morbidity. At present, chemotherapy is one of the most effective treatments; however it often fails to meet the requirements in the clinical therapy. In the present study, we have successfully formulated ligand-decorated cancer-targeted CDDP-loaded PLGA-PEG/NR7 nanoparticles and demonstrated the feasibility of using NR7 peptide for targeted delivery, rapid intracellular uptake, and enhanced cytotoxic effect in receptor-overexpressed OSCC cancer cells.
Results Nanosized particles were formed and sustained release patterns were observed for PLGA/NR7 nanoparticles. Significantly higher cellular uptake was observed in HN6 OSCC cancer cells and superior anticancer effects are observed from the optimized targeted nanoparticles. Furthermore, Live/Dead assay showed a higher extent of red fluorescence was observed for the cells exposed with PLGA/NR7 than compared with non-targeted PLGA NP. The presence of the NR7-targeting moiety on the surface of PLGA carriers could allow the specific receptor-mediated internalization, enhanced cellular uptake, and higher cell killing potency. Especially, PLGA/NR7 NP exhibited a superior apoptosis effect in HN6 cancer cells with around ~45 % (early and late apoptotic stage) and ~59 % after 24 and 48 h incubation, respectively. It is apparent that the actively targeted micelles will deliver more anticancer agent to cancer cell than non-targeted one. Conclusion Altogether, our results show the feasibility and promise of a cell-targeted anticancer nanomedicine strategy that can be effective for the treatment of oral squamous cell carcinoma. The present work might be of great importance to the further exploration of the potential application of PLGA/NR7 in the clinically relevant animal models.
Collapse
Affiliation(s)
- Zhi-Qi Wang
- Department of Head and Neck Surgery, Shandong Cancer Hospital and Institute, Jinan, 250117, China.
| | - Kai Liu
- Department of Gastrointestinal Surgery, Shandong Cancer Hospital and Institute, Jinan, 250117, China.
| | - Zhi-Jun Huo
- Department of Breast Disease Center, Shandong Cancer Hospital and Institute, Jinan, 250117, China.
| | - Xiao-Chen Li
- Department of Internal Medicine, Affiliated Hospital of Shandong Academy of Medical Sciences, Jinan, 250031, China.
| | - Min Wang
- Department of Pathology, The Second People's Hospital of Liaocheng, Linqing, 252600, Shandong, China.
| | - Ping Liu
- Department of Pharmacy, Shandong Provincial Hospital Affiliated to Shandong University, Jinan, 250021, China.
| | - Bo Pang
- Department of Neurosurgery, Qilu Hospital, Shandong Univeristy, 107# Wenhua Xi Road, Jinan, 250012, China.
| | - Shi-Jiang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Jinan, 250117, Shandong, China.
| |
Collapse
|
25
|
IND-directed safety and biodistribution study of intravenously injected cetuximab-IRDye800 in cynomolgus macaques. Mol Imaging Biol 2015; 17:49-57. [PMID: 25080323 DOI: 10.1007/s11307-014-0773-9] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
PURPOSE The use of receptor-targeted antibodies conjugated to fluorophores is actively being explored for real-time imaging of disease states; however, the toxicity of the bioconjugate has not been assessed in non-human primates. PROCEDURES To this end, the in vivo toxicity and pharmacokinetics of IRDye800 conjugated to cetuximab (cetuximab-IRDye800; 21 mg/kg; equivalent to 250 mg/m(2) human dose) were assessed in male cynomolgus monkeys over 15 days following intravenous injection and compared with an unlabeled cetuximab-dosed control group. RESULTS Cetuximab-IRDye800 was well tolerated. There were no infusion reactions, adverse clinical signs, mortality, weight loss, or clinical histopathology findings. The plasma half-life for the cetuximab-IRDye800 and cetuximab groups was equivalent (2.5 days). The total recovered cetuximab-IRDye800 in all tissues at study termination was estimated to be 12 % of the total dose. Both cetuximab-IRDye800 and cetuximab groups showed increased QTc after dosing. The QTc for the cetuximab-dosed group returned to baseline by day 15, while the QTc of the cetuximab-IRDye800 remained elevated compared to baseline. CONCLUSION IRDye800 in low molar ratios does not significantly impact cetuximab half-life or result in organ toxicity. These studies support careful cardiac monitoring (ECG) for human studies using fluorescent dyes.
Collapse
|
26
|
Warram JM, de Boer E, Korb M, Hartman Y, Kovar J, Markert JM, Gillespie GY, Rosenthal EL. Fluorescence-guided resection of experimental malignant glioma using cetuximab-IRDye 800CW. Br J Neurosurg 2015; 29:850-8. [PMID: 26073144 DOI: 10.3109/02688697.2015.1056090] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The standard treatment for glioblastoma multiforme (GBM) remains maximal safe surgical resection. Here, we evaluated the ability of a systemically administered antibody-dye probe conjugate (cetuximab-IRDye 800CW) to provide sufficient fluorescent contrast for surgical resection of disease in both subcutaneous and orthotopic animal models of GBM. Multiple luciferase-positive GBM cell lines (D-54MG, U-87MG, and U-251MG; n = 5) were implanted in mouse flank and tumors were fluorescently imaged daily using a closed-field near-infrared (NIR) system after cetuximab-IRDye 800CW systemic administration. Orthotopic models were also generated (n = 5), and tumor resection was performed under white light and fluorescence guidance using an FDA-approved wide-field NIR imaging system. Residual tumor was monitored using luciferase imaging. Immunohistochemistry was performed to characterize tumor fluorescence, epidermal growth factor receptor (EGFR) expression, and vessel density. Daily imaging of tumors revealed an average tumor-to-background (TBR) of 4.5 for U-87MG, 4.1 for D-54MG, and 3.7 for U-251MG. Fluorescence intensity within the tumors peaked on day-1 after cetuximab-IRDye 800CW administration, however the TBR increased over time in two of the three cell lines. For the orthotopic model, TBR on surgery day ranged from 19 to 23 during wide-field, intraoperative imaging. Surgical resection under white light on day 3 after cetuximab-IRDye 800CW resulted in an average 41% reduction in luciferase signal while fluorescence-guided resection using wide-field NIR imaging resulted in a significantly (P = 0.001) greater reduction in luciferase signal (87%). Reduction of luciferase signal was found to correlate (R (2) = 0.99) with reduction in fluorescence intensity. Fluorescence intensity was found to correlate (P < 0.05) with EGFR expression in D-54MG and U-251MG tumor types but not U-87MG. However, tumor fluorescence was found to correlate with vessel density for the U-87MG tumors. Here we show systemic administration of cetuximab-IRDye 800CW in combination with wide-field NIR imaging provided robust and specific fluorescence contrast for successful localization of disease in subcutaneous and orthotopic animal models of GBM.
Collapse
Affiliation(s)
- Jason M Warram
- a Department of Surgery , University of Alabama at Birmingham , Birmingham, Alabama , USA
| | - Esther de Boer
- a Department of Surgery , University of Alabama at Birmingham , Birmingham, Alabama , USA.,d Department of Surgery , University Medical Center Groningen, University of Groningen , the Netherlands
| | - Melissa Korb
- a Department of Surgery , University of Alabama at Birmingham , Birmingham, Alabama , USA
| | - Yolanda Hartman
- a Department of Surgery , University of Alabama at Birmingham , Birmingham, Alabama , USA
| | - Joy Kovar
- c LI-COR Biosciences , Lincoln, Nebraska , USA
| | - James M Markert
- b Department of Neurosurgery , University of Alabama at Birmingham , Birmingham, Alabama , USA
| | - G Yancey Gillespie
- b Department of Neurosurgery , University of Alabama at Birmingham , Birmingham, Alabama , USA
| | - Eben L Rosenthal
- a Department of Surgery , University of Alabama at Birmingham , Birmingham, Alabama , USA
| |
Collapse
|
27
|
Pogue BW, Paulsen KD, Hull SM, Samkoe KS, Gunn J, Hoopes J, Roberts DW, Strong TV, Draney D, Feldwisch J. Advancing Molecular-Guided Surgery through probe development and testing in a moderate cost evaluation pipeline. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2015; 9311:931112. [PMID: 25914500 PMCID: PMC4405779 DOI: 10.1117/12.2083224] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Molecular guided oncology surgery has the potential to transform the way decisions about resection are done, and can be critically important in areas such as neurosurgery where the margins of tumor relative to critical normal tissues are not readily apparent from visual or palpable guidance. Yet there are major financial barriers to advancing agents into clinical trials with commercial backing. We observe that development of these agents in the standard biological therapeutic paradigm is not viable, due to the high up front financial investment needed and the limitations in the revenue models of contrast agents for imaging. The hypothesized solution to this problem is to develop small molecular biologicals tagged with an established fluorescent reporter, through the chemical agent approval pathway, targeting a phase 0 trials initially, such that the initial startup phase can be completely funded by a single NIH grant. In this way, fast trials can be completed to de-risk the development pipeline, and advance the idea of fluorescence-guided surgery (FGS) reporters into human testing. As with biological therapies the potential successes of each agent are still moderate, but this process will allow the field to advance in a more stable and productive manner, rather than relying upon isolated molecules developed at high cost and risk. The pathway proposed and tested here uses peptide synthesis of an epidermal growth factor receptor (EGFR)-binding Affibody molecules, uniquely conjugated to IRDye 800CW, developed and tested in academic and industrial laboratories with well-established records for GMP production, fill & finish, toxicity testing, and early phase clinical trials with image guidance.
Collapse
Affiliation(s)
- Brian W Pogue
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA ; Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover NH 03755 USA
| | - Keith D Paulsen
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA ; Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover NH 03755 USA
| | - Sally M Hull
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA
| | - Kimberly S Samkoe
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA ; Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover NH 03755 USA
| | - Jason Gunn
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA
| | - Jack Hoopes
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA ; Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover NH 03755 USA
| | - David W Roberts
- Thayer School of Engineering, Dartmouth College, Hanover NH 03755 USA ; Department of Surgery, Geisel School of Medicine at Dartmouth, Hanover NH 03755 USA
| | - Theresa V Strong
- Department of Medicine, Univ. of Alabama at Birmingham School of Medicine, Birmingham AL 35294 USA
| | | | | |
Collapse
|
28
|
Preclinical comparison of near-infrared-labeled cetuximab and panitumumab for optical imaging of head and neck squamous cell carcinoma. Mol Imaging Biol 2014; 15:722-9. [PMID: 23715932 DOI: 10.1007/s11307-013-0652-9] [Citation(s) in RCA: 76] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
PURPOSE Though various targets have been proposed and evaluated, no agent has yet been investigated in a clinical setting for head and neck cancer. The present study aimed to compare two fluorescently labeled anti-epidermal growth factor receptor (EGFR) antibodies for detection of head and neck squamous cell carcinoma (HNSCC). PROCEDURES Antigen specificities and in vitro imaging of the fluorescently labeled anti-EGFR antibodies were performed. Next, immunodeficient mice (n = 22) bearing HNSCC (OSC-19 and SCC-1) tongue tumors received systemic injections of cetuximab-IRDye800CW, panitumumab-IRDye800CW, or IgG-IRDye800CW (a nonspecific control). Tumors were imaged and resected using two near-infrared imaging systems, SPY and Pearl. Fluorescent lymph nodes were also identified, and all resected tissues were sent for pathology. RESULTS Panitumumab-IRDye800CW and cetuximab-IRDye800CW had specific and high affinity binding for EGFR (K D = 0.12 and 0.31 nM, respectively). Panitumumab-IRDye800CW demonstrated a 2-fold increase in fluorescence intensity compared to cetuximab-IRDye800CW in vitro. In vivo, both fluorescently labeled antibodies produced higher tumor-to-background ratios compared to IgG-IRDye800CW. However, there was no significant difference between the two in either cell line or imaging modality (OSC-19: p = 0.08 SPY, p = 0.48 Pearl; SCC-1: p = 0.77 SPY, p = 0.59 Pearl; paired t tests). CONCLUSIONS There was no significant difference between the two fluorescently labeled anti-EGFR monoclonal antibodies in murine models of HNSCC. Both cetuximab and panitumumab can be considered suitable targeting agents for fluorescent intraoperative detection of HNSCC.
Collapse
|
29
|
Sun JY, Shen J, Thibodeaux J, Huang G, Wang Y, Gao J, Low PS, Dimitrov DS, Sumer BD. In vivo optical imaging of folate receptor-β in head and neck squamous cell carcinoma. Laryngoscope 2014; 124:E312-9. [PMID: 24448885 DOI: 10.1002/lary.24606] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Revised: 01/08/2014] [Accepted: 01/13/2014] [Indexed: 11/06/2022]
Abstract
OBJECTIVES/HYPOTHESIS Folate receptor (FR) expression, although known to be elevated in many types of cancer and inflammatory cells, has not been well characterized in head and neck squamous cell carcinoma (HNSCC). We hypothesized that tumor infiltrating inflammatory cells expressing FR-β could allow fluorescent visualization of HNSCC tumors using folate conjugated dyes even when FR expression in cancer cells is low. STUDY DESIGN Retrospective review of clinical pathologic specimens and in vivo animal study. METHODS A tissue microarray with tumor and tumor-free tissue from 22 patients with HNSCC was stained with antibodies to FR-α and FR-β. We characterized FR-β(+) cells by examining CD45, CD68, CD206, and transforming growth factor (TGF)-β expression. To investigate fluorescent imaging, mice with orthotopic tumor xenografts were imaged in vivo after intravenous injections of folate conjugated fluorescein isothiocyanate (folate-FITC) and were histologically evaluated ex vivo. RESULTS All tumor samples demonstrated significant FR-β staining and negligible FR-α staining. FR-β(+) cells found in tumors coexpressed CD68 and had increased expression of CD206 and TGF-β characteristic of tumor-associated macrophages. In the xenograft models, tumors showed strong in vivo fluorescence after folate-FITC injection in contrast to surrounding normal tissues. Histologic examination of the xenograft tissue similarly showed folate-FITC uptake in areas of inflammatory cellular infiltrate. CONCLUSIONS Although HNSCC tumor cells do not express FR, HNSCC tumors contain a significant population of FR-β-expressing macrophages. Folate conjugated fluorescent dye is able to specifically target and label tumor xenografts to permit macroscopic fluorescence imaging due to FR-β expression on the infiltrating inflammatory cells.
Collapse
Affiliation(s)
- Joel Y Sun
- Department of Otolaryngology, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Huang H, Bai YL, Yang K, Tang H, Wang YW. Optical imaging of head and neck squamous cell carcinoma in vivo using arginine-glycine-aspartic acid peptide conjugated near-infrared quantum dots. Onco Targets Ther 2013; 6:1779-87. [PMID: 24324343 PMCID: PMC3855103 DOI: 10.2147/ott.s53901] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Molecular imaging plays a key role in personalized medicine and tumor diagnosis. Quantum dots with near-infrared emission spectra demonstrate excellent tissue penetration and photostability, and have recently emerged as important tools for in vivo tumor imaging. Integrin αvβ3 has been shown to be highly and specifically expressed in endothelial cells of tumor angiogenic vessels in almost all types of tumors, and specifically binds to the peptide containing arginine-glycine-aspartic acid (RGD). In this study, we conjugated RGD with quantum dots with emission wavelength of 800 nm (QD800) to generate QD800-RGD, and used it via intravenous injection as a probe to image tumors in nude mice bearing head and neck squamous cell carcinoma (HNSCC). Twelve hours after the injection, the mice were still alive and were sacrificed to isolate tumors and ten major organs for ex vivo analysis to localize the probe in these tissues. The results showed that QD800-RGD was specifically targeted to integrin αvβ3 in vitro and in vivo, producing clear tumor fluorescence images after the intravenous injection. The tumor-to-background ratio and size of tumor image were highest within 6 hours of the injection and declined significantly at 9 hours after the injection, but there was still a clearly visible tumor image at 12 hours. The greatest amount of QD800-RGD was found in liver and spleen, followed by tumor and lung, and a weak fluorescence signal was seen in tibia. No detectable signal of QD800-RGD was found in brain, heart, kidney, testis, stomach, or intestine. Our study demonstrated that using integrin αvβ3 as target, it is possible to use intravenously injected QD800-RGD to generate high quality images of HNSCC, and the technique offers great potential in the diagnosis and personalized therapy for HNSCC.
Collapse
Affiliation(s)
- Hao Huang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | | | | | | | | |
Collapse
|
31
|
Korb ML, Hartman YE, Kovar J, Zinn KR, Bland KI, Rosenthal EL. Use of monoclonal antibody-IRDye800CW bioconjugates in the resection of breast cancer. J Surg Res 2013; 188:119-28. [PMID: 24360117 DOI: 10.1016/j.jss.2013.11.1089] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2013] [Revised: 11/04/2013] [Accepted: 11/15/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND Complete surgical resection of breast cancer is a powerful determinant of patient outcome, and failure to achieve negative margins results in reoperation in between 30% and 60% of patients. We hypothesize that repurposing Food and Drug Administration-approved antibodies as tumor-targeting diagnostic molecules can function as optical contrast agents to identify the boundaries of malignant tissue intraoperatively. MATERIALS AND METHODS The monoclonal antibodies bevacizumab, cetuximab, panitumumab, trastuzumab, and tocilizumab were covalently linked to a near-infrared fluorescence probe (IRDye800CW) and in vitro binding assays were performed to confirm ligand-specific binding. Nude mice bearing human breast cancer flank tumors were intravenously injected with the antibody-IRDye800 bioconjugates and imaged over time. Tumor resections were performed using the SPY and Pearl Impulse systems, and the presence or absence of tumor was confirmed by conventional and fluorescence histology. RESULTS Tumor was distinguishable from normal tissue using both SPY and Pearl systems, with both platforms being able to detect tumor as small as 0.5 mg. Serial surgical resections demonstrated that real-time fluorescence can differentiate subclinical segments of disease. Pathologic examination of samples by conventional and optical histology using the Odyssey scanner confirmed that the bioconjugates were specific for tumor cells and allowed accurate differentiation of malignant areas from normal tissue. CONCLUSIONS Human breast cancer tumors can be imaged in vivo with multiple optical imaging platforms using near-infrared fluorescently labeled antibodies. These data support additional preclinical investigations for improving the surgical resection of malignancies with the goal of eventual clinical translation.
Collapse
Affiliation(s)
- Melissa L Korb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Yolanda E Hartman
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Joy Kovar
- LI-COR Biosciences, Lincoln, Nebraska
| | - Kurt R Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Kirby I Bland
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama
| | - Eben L Rosenthal
- Department of Surgery, University of Alabama at Birmingham, Birmingham, Alabama.
| |
Collapse
|
32
|
Day KE, Beck LN, Deep NL, Kovar J, Zinn KR, Rosenthal EL. Fluorescently labeled therapeutic antibodies for detection of microscopic melanoma. Laryngoscope 2013; 123:2681-9. [PMID: 23616260 PMCID: PMC3758788 DOI: 10.1002/lary.24102] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2013] [Revised: 02/05/2013] [Accepted: 02/21/2013] [Indexed: 11/07/2022]
Abstract
OBJECTIVES/HYPOTHESIS Detection of microscopic disease during surgical resection of melanoma remains a significant challenge. To assess real-time optical imaging for visualization of microscopic cancer, we evaluated three US Food and Drug Administration (FDA)-approved therapeutic monoclonal antibodies. STUDY DESIGN Prospective, basic science. METHODS Melanoma cell lines (A375 and SKMEL5) were xenografted into the ears of immunodeficient mice. Bevacizumab, panitumumab, tocilizumab, or a nonspecific immunoglobin G (IgG) were covalently linked to a near-infrared (NIR) fluorescent probe (IRDye800CW) and systemically injected. Primary tumors were imaged and then resected under fluorescent guidance using the SPY (Novadaq, Toronto, Ontario, Canada), an NIR imaging system used in plastic and reconstructive surgeries to evaluate perfusion. Mice were also imaged with the Pearl Impulse small animal imager (LI-COR Biosciences, Lincoln, NE), an NIR imaging system designed for use with IRDye800CW. Postresection, small tissue fragments were fluorescently imaged and the presence of tumor subsequently confirmed by correlation with histology. RESULTS All fluorescently labeled therapeutic monoclonal antibodies could adequately delineate tumor from normal tissue based on tumor-to-background ratios (TBR) compared to IgG-IRDye800CW. On serial imaging, panitumumab achieved the highest TBRs with both SPY and Pearl (3.8 and 6.6, respectively). When used to guide resections, the antibody-dye conjugates generated TBRs in the range of 1.3 to 2.2 (average, 1.6) using the SPY and 1.9 to 6.3 (average, 2.7) using the Pearl. There was no significant difference among the antibodies with either imaging modality or cell line (one-way analysis of variance). CONCLUSIONS Our data suggest that FDA-approved antibodies may be suitable targeting agents for the intraoperative fluorescent detection of melanoma.
Collapse
Affiliation(s)
- Kristine E. Day
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Lauren N. Beck
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Nicholas L. Deep
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Joy Kovar
- LI-COR Biosciences, Lincoln, NE, 68504
| | - Kurt R Zinn
- Department of Radiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Eben L. Rosenthal
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL 35294
| |
Collapse
|
33
|
Heath CH, Deep NL, Sweeny L, Zinn KR, Rosenthal EL. Use of panitumumab-IRDye800 to image microscopic head and neck cancer in an orthotopic surgical model. Ann Surg Oncol 2012; 19:3879-87. [PMID: 22669455 PMCID: PMC3595117 DOI: 10.1245/s10434-012-2435-y] [Citation(s) in RCA: 85] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2012] [Indexed: 01/02/2023]
Abstract
BACKGROUND Fluorescence imaging hardware (SPY) has recently been developed for intraoperative assessment of blood flow via detection of probes emitting in the near-infrared (NIR) spectrum. This study sought to determine if this imaging system was capable of detecting micrometastatic head and neck squamous cell carcinoma (HNSCC) in preclinical models. METHODS A NIR fluorescent probe (IRDye800CW) was covalently linked to a monoclonal antibody targeting epidermal growth factor receptor (EGFR; panitumumab) or nonspecific IgG. HNSCC flank (SCC-1) and orthotopic (FADU and OSC19) xenografts were imaged 48-96 h after systemic injection of labeled panitumumab or IgG. The primary tumor and regional lymph nodes were dissected using fluorescence guidance with the SPY system and grossly assessed with a charge-coupled NIR system (Pearl). Histologic slides were also imaged with a NIR charged-coupled device (Odyssey) and fluorescence intensity was correlated with pathologic confirmation of disease. RESULTS Orthotopic tongue tumors were clearly delineated from normal tissue with tumor-to-background ratios of 2.9 (Pearl) and 2.3 (SPY). Disease detection was significantly improved with panitumumab-IRDye compared to IgG-IRDye800 (P < 0.05). Tissue biopsy samples (average size 3.7 mm) positive for fluorescence were confirmed for pathologic disease by histology and immunohistochemistry (n = 25 of 25). Biopsy samples of nonfluorescent tissue were proven to be negative for malignancy (n = 28 of 28). The SPY was able to detect regional lymph node metastasis (<1.0 mm) and microscopic areas of disease. Standard histological assessment in both frozen and paraffin-embedded histologic specimens was augmented using the Odyssey. CONCLUSIONS Panitumumab-IRDye800 may have clinical utility in detection and removal of microscopic HNSCC using existing intraoperative optical imaging hardware and may augment analysis of frozen and permanent pathology.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/surgery
- ErbB Receptors/antagonists & inhibitors
- ErbB Receptors/immunology
- Head and Neck Neoplasms/diagnosis
- Head and Neck Neoplasms/surgery
- Humans
- Immunoenzyme Techniques
- Indoles
- Lymphatic Metastasis
- Mice
- Mice, SCID
- Microscopy, Fluorescence
- Models, Anatomic
- Optical Imaging
- Panitumumab
- Spectroscopy, Near-Infrared
- Surgery, Computer-Assisted
- Transplantation, Heterologous
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- C. Hope Heath
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| | - Nicholas L. Deep
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| | - Larissa Sweeny
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| | - Kurt R Zinn
- Radiology, University of Alabama at Birmingham, Birmingham, AL 35294
| | - Eben L. Rosenthal
- Department of Surgery, University of Alabama at Birmingham,
Birmingham, AL 35294
| |
Collapse
|
34
|
Zhang L, Wang K, Zhao F, Hu W, Chen J, Lanza GM, Shen B, Zhang B. Near infrared imaging of EGFR of oral squamous cell carcinoma in mice administered arsenic trioxide. PLoS One 2012; 7:e46255. [PMID: 23029451 PMCID: PMC3460885 DOI: 10.1371/journal.pone.0046255] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2012] [Accepted: 08/31/2012] [Indexed: 02/03/2023] Open
Abstract
Background The effectiveness of near-infrared imaging (NIR) interrogation of epidermal growth factor receptor (EGFR) expression as a sensitive biomarker of oral squamous cell carcinoma (OSCC) response to arsenic trioxide therapy was studied in mice. Material and Methods A431 OSCC in vitro were exposed to 0 µM, 0.5 µM, 2.5 µM, or 5 µM of As2O3 for 0 h, 24 h, 48 h and 72 h. Confocal microscopy and flow cytometry confirmed EGFR expression and demonstrated a sensitivity dose-related signal decline with As2O3 treatment. Next, mice with pharynx-implanted A431 cells received As2O3 i.p. every 48 h at 0.0, 0.5, 2.5, or 5 mg/kg/day (n = 6/group) from day 0 to 10. An intravenous NIR probe, EGF-Cy5.5, was injected at baseline and on days 4, 8, and 12 for dynamic NIR imaging. Tumor volume and body weights were measured three times weekly. Results In vitro, A431 EGFR expression was well appreciated in the controls and decreased (p<0.05) with increasing As2O3 dose and treatment duration. In vivo EGFR NIR tumor signal intensity decreased (p<0.05) in As2O3 treated groups versus controls from days 4 to 12, consistent with increasing dosage. Tumor volume diminished in a dose-related manner while body weight was unaffected. Immunohistochemical staining of excised tumors confirmed that EGFR expression was reduced by As2O3 treatment in a dose responsive pattern. Conclusion This study demonstrates for the first time that OSCC can be interrogated in vivo by NIR molecular imaging of the EGFR and that this biomarker is effective for the longitudinal assessment of OSCC response to As2O3 treatment.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Arsenic Trioxide
- Arsenicals/pharmacology
- Arsenicals/therapeutic use
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Body Weight/drug effects
- Carbocyanines
- Carcinoma, Squamous Cell/diagnosis
- Carcinoma, Squamous Cell/drug therapy
- Carcinoma, Squamous Cell/genetics
- Carcinoma, Squamous Cell/metabolism
- Dose-Response Relationship, Drug
- Epidermal Growth Factor/administration & dosage
- Epidermal Growth Factor/chemistry
- ErbB Receptors/analysis
- ErbB Receptors/genetics
- ErbB Receptors/metabolism
- Gene Expression/drug effects
- Injections, Intraperitoneal
- Injections, Intravenous
- Magnetic Resonance Imaging
- Mice
- Mice, Nude
- Mouth Neoplasms/diagnosis
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/genetics
- Mouth Neoplasms/metabolism
- Neoplasms, Experimental
- Oxides/pharmacology
- Oxides/therapeutic use
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/chemistry
- Spectroscopy, Near-Infrared
- Tumor Burden/drug effects
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Lingbo Zhang
- Stomatology Department, Institute of Hard Tissue Development and Regeneration, 2nd Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Kezheng Wang
- Radiology Department and Molecular Imaging Center, 4th Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Falin Zhao
- School of Health Management, Hangzhou Normal University, Hangzhou, Zhejiang, China
| | - Weiping Hu
- Stomatology Department, Institute of Hard Tissue Development and Regeneration, 2nd Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
| | - Junjie Chen
- Division of Cardiology and C-TRAIN, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Gregory M. Lanza
- Division of Cardiology and C-TRAIN, Washington University School of Medicine, St. Louis, Missouri, United States of America
| | - Baozhong Shen
- Radiology Department and Molecular Imaging Center, 4th Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
- * E-mail: (BS); (BZ)
| | - Bin Zhang
- Stomatology Department, Institute of Hard Tissue Development and Regeneration, 2nd Affiliated Hospital, Harbin Medical University, Harbin, Heilongjiang, China
- * E-mail: (BS); (BZ)
| |
Collapse
|
35
|
Evaluation of tyrosine receptor kinases in the interactions of head and neck squamous cell carcinoma cells and fibroblasts. Oral Oncol 2012; 48:1242-9. [PMID: 22795534 DOI: 10.1016/j.oraloncology.2012.06.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2012] [Revised: 06/12/2012] [Accepted: 06/17/2012] [Indexed: 11/24/2022]
Abstract
OBJECTIVE Despite treatment advancements, disease-free survival of head and neck squamous cell carcinoma (HNSCC) has not significantly improved. This may be a result of tumor-fibroblasts interactions providing protective pathways for oncogenic cells to resist therapy. Further understanding of these relationships in HNSCC may improve effectiveness of targeted therapies. In this article, we investigated the role of several receptor tyrosine kinases (RTKs) in the interactions between HNSCC cells and supporting cells (fibroblasts). MATERIALS AND METHODS HNSCC cell lines and human tumor samples were evaluated for FGFR1/2/3, and PDGF-beta expression levels. Cell lines (FADU, SCC1, OSC19, Cal27, SCC22A) were treated with a range of physiological concentrations of dovitinib and assessed for proliferation, cytotoxicity, and apoptosis. Mice bearing HNSCC xenografts were treated with dovitinib (20 mg/kg). RESULTS Evaluation of HNSCC tumor specimens, cell lines and fibroblasts found variable expression of multiple RTKs (fibroblasts growth factor receptor, platelet derived growth factor receptor and vascular endothelial growth factor receptor) and their ligands, supporting previous theories of paracrine and autocrine signaling within the microenvironment. In a dose-dependent fashion, RTK inhibition reduced proliferation of HNSCC cell lines and fibroblast in vitro. When HNSCC cells were cocultured with fibroblasts, RTK inhibition resulted in a smaller reduction in the proliferation relative to untreated conditions. In vivo, RTK inhibition resulted in significant tumor regression and growth inhibition (p<0.05) and reduced the incidence of regional lymph node metastasis. CONCLUSION Effective treatment of HNSCC, therefore, may require inhibition of multiple RTKs in order to adequately inhibit the microenvironment's various signaling pathways.
Collapse
|
36
|
Cohen-Kaplan V, Jrbashyan J, Yanir Y, Naroditsky I, Ben-Izhak O, Ilan N, Doweck I, Vlodavsky I. Heparanase induces signal transducer and activator of transcription (STAT) protein phosphorylation: preclinical and clinical significance in head and neck cancer. J Biol Chem 2011; 287:6668-78. [PMID: 22194600 DOI: 10.1074/jbc.m111.271346] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Activity of heparanase is implicated strongly in dissemination of metastatic tumor cells and cells of the immune system. In addition, heparanase enhances the phosphorylation of selected signaling molecules, including SRC and EGFR, in a manner that requires secretion but not enzymatic activity of heparanase and is mediated by its C-terminal domain. Clinically, heparanase staining is associated with larger tumors and increased EGFR phosphorylation in head and neck carcinoma. We hypothesized that signal transducer and activator of transcription (STAT) proteins mediate the protumorigenic function of heparanase downstream of the EGFR. We provide evidence that heparanase enhances the phosphorylation of STAT3 and STAT5b but not STAT5a. Moreover, enhanced proliferation of heparanase transfected cells was attenuated by STAT3 and STAT5b siRNA, but not STAT5a or STAT1 siRNA. Clinically, STAT3 phosphorylation was associated with head and neck cancer progression, EGFR phosphorylation, and heparanase expression and cellular localization. Notably, cytoplasmic rather than nuclear phospho-STAT3 correlated with increased tumor size (T-stage; p = 0.007), number of metastatic neck lymph nodes (p = 0.05), and reduced survival of patients (p = 0.04).
Collapse
Affiliation(s)
- Victoria Cohen-Kaplan
- Cancer and Vascular Biology Research Center, Bruce Rappaport Faculty of Medicine, Technion, Haifa 31096, Israel
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Miyamoto S, Sperry S, Yamashita T, Reddy NP, O'Malley BW, Li D. Molecular imaging assisted surgery improves survival in a murine head and neck cancer model. Int J Cancer 2011; 131:1235-42. [DOI: 10.1002/ijc.27328] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2011] [Accepted: 10/05/2011] [Indexed: 01/03/2023]
|
38
|
Kaur S, Venktaraman G, Jain M, Senapati S, Garg PK, Batra SK. Recent trends in antibody-based oncologic imaging. Cancer Lett 2011; 315:97-111. [PMID: 22104729 DOI: 10.1016/j.canlet.2011.10.017] [Citation(s) in RCA: 80] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2011] [Revised: 10/11/2011] [Accepted: 10/12/2011] [Indexed: 01/27/2023]
Abstract
Antibodies, with their unmatched ability for selective binding to any target, are considered as potentially the most specific probes for imaging. Their clinical utility, however, has been limited chiefly due to their slow clearance from the circulation, longer retention in non-targeted tissues and the extensive optimization required for each antibody-tracer. The development of newer contrast agents, combined with improved conjugation strategies and novel engineered forms of antibodies (diabodies, minibodies, single chain variable fragments, and nanobodies), have triggered a new wave of antibody-based imaging approaches. Apart from their conventional use with nuclear imaging probes, antibodies and their modified forms are increasingly being employed with non-radioisotopic contrast agents (MRI and ultrasound) as well as newer imaging modalities, such as quantum dots, near infra red (NIR) probes, nanoshells and surface enhanced Raman spectroscopy (SERS). The review article discusses new developments in the usage of antibodies and their modified forms in conjunction with probes of various imaging modalities such as nuclear imaging, optical imaging, ultrasound, MRI, SERS and nanoshells in preclinical and clinical studies on the diagnosis, prognosis and therapeutic responses of cancer.
Collapse
Affiliation(s)
- Sukhwinder Kaur
- Department of Biochemistry and Molecular Biology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | | | | | | | | | | |
Collapse
|
39
|
Keereweer S, Kerrebijn JDF, Mol IM, Mieog JSD, Van Driel PBAA, Baatenburg de Jong RJ, Vahrmeijer AL, Löwik CWGM. Optical imaging of oral squamous cell carcinoma and cervical lymph node metastasis. Head Neck 2011; 34:1002-8. [PMID: 21987435 DOI: 10.1002/hed.21861] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/20/2011] [Indexed: 12/22/2022] Open
Abstract
BACKGROUND In oral cancer surgery, intraoperative optical imaging could help the surgeon to determine adequate tumor-free margins. METHODS Tumor-specific near-infrared fluorescence agents targeting epidermal growth factor receptor (CW800 EGF) or glucose transporter system (CW800 2-DG) were administered to mice with tongue carcinoma and cervical lymph node metastases. Tumor growth was followed by bioluminescence imaging. Fluorescence signals were compared with a control group of healthy animals. RESULTS Significantly higher fluorescence was found in tongue tumors and cervical lymph node metastases compared with that in control animals. Fluorescence correlated with histopathology. Tumor-to-background ratio of CW800 EGF in the tongue was 13.8 (SD = 6.1) and in the lymph nodes 15.7 (SD = 8.8). For CW800 2-DG, the tumor-to-background ratio in the tongue was 4.6 (SD = 2.1) and in the lymph nodes 33.9 (SD = 18.4). CONCLUSIONS Optical imaging can be used to detect oral cancer and cervical lymph node metastases and could potentially improve complete surgical resection by real-time image-guided surgery.
Collapse
Affiliation(s)
- Stijn Keereweer
- Erasmus Medical Center, Department of Otorhinolaryngology Head and Neck Surgery, Rotterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Yang K, Zhang FJ, Tang H, Zhao C, Cao YA, Lv XQ, Chen D, Li YD. In-vivo imaging of oral squamous cell carcinoma by EGFR monoclonal antibody conjugated near-infrared quantum dots in mice. Int J Nanomedicine 2011; 6:1739-45. [PMID: 21980236 PMCID: PMC3184933 DOI: 10.2147/ijn.s23348] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2011] [Indexed: 01/17/2023] Open
Abstract
Objectives: The purpose of this study was to investigate in-vivo visible imaging of oral squamous cell carcinoma (OSCC) by targeting epidermal growth factor receptor (EGFR) with near-infrared quantum dots. Materials and methods: Quantum dots with an emission wavelength of 800 nm (QD800) were conjugated to monoclonal antibodies against EGFR, resulting in the probe designated as QD800-EGFR Ab. OSCC cell line (BcaCD885) expressing high levels of EGFR was transplanted subcutaneously into nude mice cheeks to develop an OSCC animal model. QD800-EGFR Ab containing 100 pmol equivalent of QD800 was intravenously injected into the animal model, and in-situ and in-vivo imaging of cheek squamous cell carcinoma was analyzed at 10 different time points. Results and conclusion: In-vivo imaging and immunohistochemical examination of the tumors showed that intravenously injected QD800-EGFR Ab probe could bind EGFR expressed on BcaCD885 cells. Fluorescence signals of BcaCD885 cells labeled with QD800-EGFR Ab probe could be clearly detected, and these fluorescence signals lasted for 24 hours. The most complete tumor images with maximal signal-to-noise ratio were observed from 15 minutes to 6 hours after injection of the probe. To the best of the authors’ knowledge, this is the first study that has obtained clear in-situ and in-vivo imaging of head and neck cancer by using QD800-EGFR Ab probe. The authors conclude that the combination of near-infrared quantum dots that are highly penetrating for tissues with EGFR monoclonal antibody has promising prospects in in-vivo imaging of OSCC and development of personalized surgical therapies.
Collapse
Affiliation(s)
- Kai Yang
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital, Chongqing Medical University, Chongqing, China.
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Acute vascular disruption by 5,6-dimethylxanthenone-4-acetic Acid in an orthotopic model of human head and neck cancer. Transl Oncol 2011; 2:121-7. [PMID: 19701496 DOI: 10.1593/tlo.09103] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2009] [Revised: 03/24/2009] [Accepted: 04/07/2009] [Indexed: 11/18/2022] Open
Abstract
The sustenance of most solid tumors including head and neck cancers (HNCs) is strongly dependent on the presence of a functioning vascular network. In this study, we examined the acute effects of a tumor vascular disrupting agent (VDA), 5,6-dimethylxanthenone-4-acetic acid (DMXAA; ASA404), in an orthotopic model of human HNC. Noninvasive magnetic resonance imaging (MRI) was used to monitor the vascular response of orthotopic FaDu xenografts to VDA therapy. Untreated tumors showed a marked but heterogeneous pattern of enhancement after contrast agent injection on serial T1-weighted (T1W) MR images. After VDA treatment, T2W and T1W MRI revealed evidence of hemorrhaging and lack of functioning vessels (enhancement) within the tumor. Quantitative estimates of relative vascular volume also showed a significant (P < .01) reduction in DMXAA-treated tumors 24 hours after therapy compared with untreated controls. Histology and immunostaining of untreated orthotopic FaDu tumors revealed poorly differentiated squamous cell carcinoma histology with distinctly visible CD31(+) endothelial cells. In sharp contrast, minimal CD31 staining with irregular endothelial fragments and faint outlines of blood vessels were seen in DMXAA-treated tumor sections. CD31 immunostaining and histology also highlighted the selectivity of vascular damage and tissue necrosis after VDA therapy with no evidence of toxicity observed in normal salivary gland, heart, liver, and skeletal muscle tissues. Together, our results demonstrate a potent and selective vascular disruptive activity of DMXAA in an orthotopic HNC model. Further evaluation into its antitumor effects alone and in combination with other agents is warranted.
Collapse
|
42
|
Reddy NP, Miyamoto S, Araki K, Liu T, Feldman M, O'Malley BW, Li D. A novel orthotopic mouse model of head and neck cancer with molecular imaging. Laryngoscope 2011; 121:1202-7. [PMID: 21480280 DOI: 10.1002/lary.21794] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2010] [Accepted: 01/21/2011] [Indexed: 11/06/2022]
Abstract
OBJECTIVES/HYPOTHESIS Our goal was to develop a noninvasive, dynamic imaging method that would further the understanding of head and neck cancer (HNC) tumor growth and local spreading. We developed a novel orthotopic mouse model of HNC with a stable cell line expressing a red fluorescent protein gene to compare a molecular imaging tumor quantification with traditional caliper measurement. METHODS An HNC-tdT stable cell line expressing the tdTomato gene was established, which were injected into the floor of the mouth of nude mice. Tumor growth was constantly monitored using molecular imaging for up to 35 days. The tumors were further evaluated by histologic examination. RESULTS Established tumors consistently expressed fluorescent signals that were successfully imaged by molecular imaging during the study. Initial tumor development was detected earlier than caliper measurement would allow. The fluorescent signal quantities of tumors detected by the imaging correlated with the tumor sizes measured by calipers. CONCLUSIONS This novel animal model represents an orthotopic human HNC model. The tumor can be detected earlier with molecular imaging than by conventional external caliper measurement. Unlike surgical measurement, the tumor can be quantified without disturbing the tumor environment. This model has significant potential for HNC oncologic research.
Collapse
Affiliation(s)
- Nishant P Reddy
- Department of Otorhinolaryngology-Head & Neck Surgery, University of Pennsylvania, Philadelphia, Pennsylvania 19104, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Keereweer S, Sterenborg HJCM, Kerrebijn JDF, Van Driel PBAA, Baatenburg de Jong RJ, Löwik CWGM. Image-guided surgery in head and neck cancer: current practice and future directions of optical imaging. Head Neck 2011; 34:120-6. [PMID: 21284051 DOI: 10.1002/hed.21625] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2010] [Accepted: 08/12/2010] [Indexed: 11/08/2022] Open
Abstract
A key aspect for the postoperative prognosis of patients with head and neck cancer is complete tumor resection. In current practice, the intraoperative assessment of the tumor-free margin is dependent on visual appearance and palpation of the tumor. Optical imaging has the potential of traversing the gap between radiology and surgery by providing real-time visualization of the tumor, thereby allowing for image-guided surgery. The use of the near-infrared light spectrum offers 2 essential advantages: increased tissue penetration of light and an increased signal-to-background ratio of contrast agents. In this review, the current practice and limitations of image-guided surgery by optical imaging using intrinsic fluorescence or contrast agents are described. Furthermore, we provide an overview of the various molecular contrast agents targeting specific hallmarks of cancer that have been used in other fields of oncologic surgery, and we describe perspectives on its future use in head and neck cancer surgery.
Collapse
Affiliation(s)
- S Keereweer
- Department of Otorhinolaryngology-Head and Neck Surgery, Erasmus Medical Center, Rotterdam, The Netherlands.
| | | | | | | | | | | |
Collapse
|
44
|
Yang K, Cao YA, Shi C, Li ZG, Zhang FJ, Yang J, Zhao C. Quantum dot-based visual in vivo imaging for oral squamous cell carcinoma in mice. Oral Oncol 2010; 46:864-8. [DOI: 10.1016/j.oraloncology.2010.09.009] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2010] [Revised: 09/21/2010] [Accepted: 09/22/2010] [Indexed: 11/28/2022]
|
45
|
Villaraza AJL, Milenic DE, Brechbiel MW. Improved speciation characteristics of PEGylated indocyanine green-labeled Panitumumab: revisiting the solution and spectroscopic properties of a near-infrared emitting anti-HER1 antibody for optical imaging of cancer. Bioconjug Chem 2010; 21:2305-12. [PMID: 21073171 DOI: 10.1021/bc100336b] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
A water-soluble amine-reactive PEGylated analogue of near-infrared emitting dye indocyanine green (5) was synthesized and used to label the anti-HER1 antibody panitumumab (Vectibix) at various equivalents. These conjugates were compared with non-PEGylated analogue conjugate products and the solution speciation analyzed with UV-vis spectrophotometry, size exclusion HPLC, and SDS-PAGE. PEGylation of the bioconjugates was successful in preventing aggregation in solution, a phenomenon observed with the non-PEGylated bioconjugates presumably due to the hydrophobicity of indocyanine green. Competitive radioimmunoassay demonstrated that the targeting moiety of the PEGylated bioconjugates was conserved. Fluorescence microscopy also demonstrated membrane binding of the bioconjugate to HER1-expressing A431 cells. Hence, these bioconjugates are suitable candidates for the in vivo optical imaging of HER1-expressing tumors.
Collapse
Affiliation(s)
- Aaron Joseph L Villaraza
- Radioimmune and Inorganic Chemistry Section, Radiation Oncology Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892-1002, USA
| | | | | |
Collapse
|
46
|
Liao C, Sun Q, Liang B, Shen J, Shuai X. Targeting EGFR-overexpressing tumor cells using Cetuximab-immunomicelles loaded with doxorubicin and superparamagnetic iron oxide. Eur J Radiol 2010; 80:699-705. [PMID: 20810233 DOI: 10.1016/j.ejrad.2010.08.005] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2010] [Revised: 07/26/2010] [Accepted: 08/02/2010] [Indexed: 01/01/2023]
Abstract
Epidermal growth factor receptor (EGFR), a cellular transmembrane receptor, plays a key role in cell proliferation and is linked to a poor prognosis in various human cancers. In this study, we constructed Cetuximab-immunomicelles in which the anti-EGFR monoclonal antibody was linked to poly(ethylene glycol)-block-poly(ɛ-caprolactone) (PEG-PCL) nanomicelles that were loaded with doxorubicin (DOX) and superparamagnetic iron oxide (SPIO). The specific interactions between EGFR-overexpressing tumor cells (A431) and immunomicelles were observed using confocal laser scanning microscopy (CLSM) and flow cytometry. Furthermore, the capacity of transporting SPIO into tumor cells using these immunomicelles was evaluated with a 1.5 T clinical magnetic resonance imaging (MRI) scanner. It was found that the acquired MRI T2 signal intensity of A431 cells that were treated with the SPIO-loaded and antibody-functionalized micelles decreased significantly. Using the thiazolyl blue tetrazolium bromide (MTT) assay, we also demonstrated that the immunomicelles inhibited cell proliferation more effectively than their nontargeting counterparts. Our results suggest that Cetuximab-immunomicelles are a useful delivery vehicle for DOX and SPIO to EGFR-overexpressing tumor cells in vitro and that Cetuximab-immunomicelles can serve as a MRI-visible and targeted drug delivery agent for better tumor imaging and therapy.
Collapse
Affiliation(s)
- Chengde Liao
- Department of Radiology, The Second Affiliated Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | | | | | | | | |
Collapse
|
47
|
Zou P, Xu S, Povoski SP, Wang A, Johnson MA, Martin EW, Subramaniam V, Xu R, Sun D. Near-infrared fluorescence labeled anti-TAG-72 monoclonal antibodies for tumor imaging in colorectal cancer xenograft mice. Mol Pharm 2009; 6:428-40. [PMID: 19718796 DOI: 10.1021/mp9000052] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Anti-TAG-72 monoclonal antibodies target the tumor-associated glycoprotein (TAG)-72 in various solid tumors. This study evaluated the use of anti-TAG-72 monoclonal antibodies, both murine CC49 and humanized CC49 (HuCC49deltaCH2), for near-infrared fluorescent (NIR) tumor imaging in colorectal cancer xenograft models. The murine CC49 and HuCC49deltaCH2 were conjugated with Cy7 monofunctional N-hydroxysuccinimide ester (Cy7-NHS). Both in vitro and in vivo anti-TAG-72 antibody binding studies were performed. The in vitro study utilized the human colon adenocarcinoma cell line LS174T that was incubated with Cy7, antibody-Cy7 conjugates, or excessive murine CC49 followed by the antibody-Cy7 conjugates and was imaged by fluorescence microscopy. The in vivo study utilized xenograft mice, bearing LS174T subcutaneous tumor implants, that received tail vein injections of Cy7, murine CC49-Cy7, HuCC49deltaCH2-Cy7, or nonspecific IgG-Cy7 and were imaged by the Xenogen IVIS 100 system from 15 min to 288 h. The biodistribution of the fluorescence labeled antibodies was determined by imaging the dissected tissues. The in vitro study revealed that the antibody-Cy7 conjugates bound to LS174T cells and were blocked by excessive murine CC49. The in vivo study demonstrated that murine CC49 achieved a tumor/blood ratio of 15 at 96 h postinjection. In comparison, HuCC49deltaCH2-Cy7 cleared much faster than murine CC49-Cy7 from the xenograft mice, and HuCC49deltaCH2-Cy7 achieved a tumor/blood ratio of 12 at 18 h postinjection. In contrast, Cy7 and Cy7 labeled nonspecific IgG resulted in no demonstrable tumor accumulation. When mice were injected with excessive unlabeled murine CC49 at 6 h before the injection of murine CC49-Cy7 or HuCC49deltaCH2-Cy7, both the intensity and retention time of the fluorescence from the tumor were reduced. In summary, the Cy7 labeled murine CC49 and HuCC49deltaCH2 demonstrate tumor-targeting capabilities in living colorectal cancer xenograft mice and provide an alternative modality for tumor imaging.
Collapse
Affiliation(s)
- Peng Zou
- Department of Pharmaceutical Sciences, College of Pharmacy, The University of Michigan, 428 Church Street, Ann Arbor, Michigan 48109, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Devaraj N, Upadhyay R, Haun J, Hilderbrand S, Weissleder R. Fast and Sensitive Pretargeted Labeling of Cancer Cells through a Tetrazine/trans-Cyclooctene Cycloaddition. Angew Chem Int Ed Engl 2009. [DOI: 10.1002/ange.200903233] [Citation(s) in RCA: 75] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
49
|
Levashova Z, Backer MV, Horng G, Felsher D, Backer JM, Blankenberg FG. SPECT and PET Imaging of EGF Receptors with Site-Specifically Labeled EGF and Dimeric EGF. Bioconjug Chem 2009; 20:742-9. [DOI: 10.1021/bc800443w] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Affiliation(s)
- Zoya Levashova
- Department of Radiology/MIPS, Stanford University School of Medicine, Stanford, California 94305, Sibtech, Inc., Brookfield, Connecticut 06804, and Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California 94305
| | - Marina V. Backer
- Department of Radiology/MIPS, Stanford University School of Medicine, Stanford, California 94305, Sibtech, Inc., Brookfield, Connecticut 06804, and Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California 94305
| | - George Horng
- Department of Radiology/MIPS, Stanford University School of Medicine, Stanford, California 94305, Sibtech, Inc., Brookfield, Connecticut 06804, and Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California 94305
| | - Dean Felsher
- Department of Radiology/MIPS, Stanford University School of Medicine, Stanford, California 94305, Sibtech, Inc., Brookfield, Connecticut 06804, and Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California 94305
| | - Joseph M. Backer
- Department of Radiology/MIPS, Stanford University School of Medicine, Stanford, California 94305, Sibtech, Inc., Brookfield, Connecticut 06804, and Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California 94305
| | - Francis G. Blankenberg
- Department of Radiology/MIPS, Stanford University School of Medicine, Stanford, California 94305, Sibtech, Inc., Brookfield, Connecticut 06804, and Department of Medicine/Oncology, Stanford University School of Medicine, Stanford, California 94305
| |
Collapse
|
50
|
Fux L, Feibish N, Cohen-Kaplan V, Gingis-Velitski S, Feld S, Geffen C, Vlodavsky I, Ilan N. Structure-function approach identifies a COOH-terminal domain that mediates heparanase signaling. Cancer Res 2009; 69:1758-67. [PMID: 19244131 DOI: 10.1158/0008-5472.can-08-1837] [Citation(s) in RCA: 80] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Heparanase is an endo-beta-d-glucuronidase capable of cleaving heparan sulfate, activity that is strongly implicated in cellular invasion associated with tumor metastasis, angiogenesis, and inflammation. In addition, heparanase was noted to exert biological functions apparently independent of its enzymatic activity, enhancing the phosphorylation of selected protein kinases and inducing gene transcription. A predicted three-dimensional structure of constitutively active heparanase clearly delineates a TIM-barrel fold previously anticipated for the enzyme. Interestingly, the model also revealed the existence of a COOH-terminal domain (C-domain) that apparently is not an integral part of the TIM-barrel fold. We provide evidence that the C-domain is critical for heparanase enzymatic activity and secretion. Moreover, the C-domain was found to mediate nonenzymatic functions of heparanase, facilitating Akt phosphorylation, cell proliferation, and tumor xenograft progression. These findings support the notion that heparanase exerts enzymatic activity-independent functions, and identify, for the first time, a protein domain responsible for heparanase-mediated signaling. Inhibitors directed against the C-domain, combined with inhibitors of heparanase enzymatic activity, are expected to neutralize heparanase functions and to profoundly affect tumor growth, angiogenesis, and metastasis.
Collapse
Affiliation(s)
- Liat Fux
- Cancer and Vascular Biology Research Center, The Bruce Rappaport Faculty of Medicine, Technion, Haifa, Israel
| | | | | | | | | | | | | | | |
Collapse
|