1
|
van Schaik JE, van der Vegt B, Slagter-Menkema L, Hanemaaijer SH, Halmos GB, Witjes MJH, van der Laan BFAM, Fehrmann RSN, Oosting SF, Plaat BEC. Potential imaging targets in primary head and neck squamous cell carcinoma and lymph node metastases. Am J Otolaryngol 2024; 45:104298. [PMID: 38640809 DOI: 10.1016/j.amjoto.2024.104298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Accepted: 04/14/2024] [Indexed: 04/21/2024]
Abstract
PURPOSE To investigate glycoprotein nonmetastatic melanoma protein B (GPNMB) and vascular endothelial growth factor (VEGF) as potential fluorescent imaging markers by comparing their protein expression to epidermal growth factor receptor (EGFR). MATERIALS AND METHODS Thirty-eight paired samples of untreated head and neck squamous cell carcinoma (HNSCC) primary tumours (PT) and corresponding synchronous lymph node metastases (LNM) were selected. After immunohistochemical staining, expression was assessed and compared by the percentage of positive tumour cells. Data were analysed using the Mann-Whitney test, effect sizes (ESr) and Spearman's correlation coefficient (r). RESULTS GPNMB expression was observed in 100 % of PT, and median 80 % (range 5-100 %) of tumour cells, VEGF in 92 % and 60 % (0-100 %), EGFR in 87 % and 60 % (0-100 %) respectively. In corresponding LNM, GPNMB expression was observed in 100 % of LNM and median 90 % (20-100 %) of tumour cells, VEGF in 87 % and 65 % (0-100 %), and EGFR in 84 % and 35 % (0-100 %). A positive correlation was found between expression in PT and LNM for GPNMB (r = 0.548) and EGFR (r = 0.618) (p < 0.001), but not for VEGF (r = -0.020; p = 0.905). GPNMB expression was present in a higher percentage of tumour cells compared to EGFR in PT (p = 0.015, ESr = -0.320) and in LNM (p < 0.001, ESr = -0.478), while VEGF was not (p = 1.00, ESr = -0.109 and - 0.152, respectively). CONCLUSION GPNMB expression is higher than EGFR in untreated HNSCC PT and corresponding LNM, while VEGF expression is comparable to EGFR. GPNMB is a promising target for fluorescent imaging in HNSCC.
Collapse
Affiliation(s)
- Jeroen E van Schaik
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Bert van der Vegt
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Lorian Slagter-Menkema
- Department of Pathology and Medical Biology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Saskia H Hanemaaijer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Gyorgi B Halmos
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Max J H Witjes
- Department of Oral & Maxillofacial Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Bernard F A M van der Laan
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Rudolf S N Fehrmann
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Sjoukje F Oosting
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands
| | - Boudewijn E C Plaat
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Groningen, University Medical Center Groningen, Hanzeplein 1, 9713 GZ Groningen, Netherlands.
| |
Collapse
|
2
|
Chatzilakou E, Hu Y, Jiang N, Yetisen AK. Biosensors for melanoma skin cancer diagnostics. Biosens Bioelectron 2024; 250:116045. [PMID: 38301546 DOI: 10.1016/j.bios.2024.116045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 01/10/2024] [Accepted: 01/15/2024] [Indexed: 02/03/2024]
Abstract
Skin cancer is a critical global public health concern, with melanoma being the deadliest variant, correlated to 80% of skin cancer-related deaths and a remarkable propensity to metastasize. Despite notable progress in skin cancer prevention and diagnosis, the limitations of existing methods accentuate the demand for precise diagnostic tools. Biosensors have emerged as valuable clinical tools, enabling rapid and reliable point-of-care (POC) testing of skin cancer. This review offers insights into skin cancer development, highlights essential cutaneous melanoma biomarkers, and assesses the current landscape of biosensing technologies for diagnosis. The comprehensive analysis in this review underscores the transformative potential of biosensors in revolutionizing melanoma skin cancer diagnosis, emphasizing their critical role in advancing patient outcomes and healthcare efficiency. The increasing availability of these approaches supports direct diagnosis and aims to reduce the reliance on biopsies, enhancing POC diagnosis. Recent advancements in biosensors for skin cancer diagnosis hold great promise, with their integration into healthcare expected to enhance early detection accuracy and reliability, thereby mitigating socioeconomic disparities.
Collapse
Affiliation(s)
- Eleni Chatzilakou
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2BU, UK
| | - Yubing Hu
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2BU, UK.
| | - Nan Jiang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China; JinFeng Laboratory, Chongqing, 401329, China.
| | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, South Kensington, London, SW7 2BU, UK.
| |
Collapse
|
3
|
Wu Y, Wu H, Lu X, Chen Y, Zhang X, Ju J, Zhang D, Zhu B, Huang S. Development and Evaluation of Targeted Optical Imaging Probes for Image‐Guided Surgery in Head and Neck Cancer. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100196] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Yue Wu
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Haiwei Wu
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Xiaoya Lu
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Yi Chen
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Xue Zhang
- University of Jinan Jinan Shandong 250021 China
| | - Jiandong Ju
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Dongsheng Zhang
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| | - Baocun Zhu
- University of Jinan Jinan Shandong 250021 China
| | - Shengyun Huang
- Department of Oral and Maxillofacial Surgery Shandong Provincial Hospital Cheeloo College of Medicine Shandong University Jinan Shandong 250021 China
| |
Collapse
|
4
|
van Driel PBAA, Keereweer S, Lowik CWGM, Oliveira S. Investigation of the Therapeutic Potential of Nanobody-Targeted Photodynamic Therapy in an Orthotopic Head and Neck Cancer Model. Methods Mol Biol 2022; 2451:521-531. [PMID: 35505029 DOI: 10.1007/978-1-0716-2099-1_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Photodynamic therapy (PDT) has a great therapeutic potential because it induces local cellular cytotoxicity upon application of a laser light that excites a photosensitizer, leading to toxic reactive oxygen species. Nevertheless, PDT still is underutilized in the clinic, mostly because of damage induced to normal surrounding tissues. Efforts have been made to improve the specificity. Nanobody-targeted PDT is one of such approaches, in which the variable domain of heavy-chain antibodies, i.e., nanobodies, are used to target photosensitizers selectively to cancer cells. In vitro studies are certainly very valuable to evaluate the therapeutic potential of PDT approaches, but many aspects such as bio-distribution of the photosensitizers, penetration through tissues, and clearance are not taken into account. In vivo studies are therefore essential to assess the influence of such factors, in order to gain more insights into the therapeutic potential of a treatment under development. This chapter describes the development of an orthotopic model of head and neck cancer, to which nanobody-targeted PDT is applied, and the therapeutic potential is assessed by immunohistochemistry one day after PDT.
Collapse
Affiliation(s)
- Pieter B A A van Driel
- Department of Orthopaedic Surgery, Leiden University Medical Center, Leiden, The Netherlands
| | - Stijn Keereweer
- Department of Otorhinolaryngology, Head and Neck Surgery, Erasmus Medical Centre, Rotterdam, The Netherlands
| | - Clemens W G M Lowik
- Department of Radiology & Nuclear Medicine, Optical Molecular Imaging, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Sabrina Oliveira
- Cell Biology, Neurobiology and Biophysics, Department of Biology, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
- Pharmaceutics, Department of Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands.
| |
Collapse
|
5
|
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
|
6
|
Stepan KO, Li MM, Kang SY, Puram SV. Molecular margins in head and neck cancer: Current techniques and future directions. Oral Oncol 2020; 110:104893. [PMID: 32702629 DOI: 10.1016/j.oraloncology.2020.104893] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2020] [Accepted: 07/01/2020] [Indexed: 12/14/2022]
Abstract
Complete tumor extirpation with clear surgical margins remains a central tenet of oncologic head and neck surgery. Rates of locoregional recurrence and survival are both significantly worse when clear margins are unable to be obtained. Current clinical practice relies on the use of frozen sections intra-operatively, followed by traditional histopathologic analysis post-operatively to assess the surgical margin. However, with improved understanding of tumor biology and advances in technology, new techniques have emerged to analyze margins at a molecular level. Such molecular margin analysis interrogates tissue for genetic, epigenetic, or proteomic changes that may belie tumor presence or aggressive features not captured by standard histopathologic techniques. Intra-operatively, this information may be used to guide resection, while post-operatively, it may help to stratify patients for adjuvant treatment. In this review, we summarize the current state of molecular margin analysis and describe directions for future research.
Collapse
Affiliation(s)
- Katelyn O Stepan
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, 4921 Parkview Pl, St. Louis, MO 63110, USA
| | - Michael M Li
- Department of Otolaryngology - Head and Neck Surgery, Ohio State University Wexner Medical Center, 410 W. 10(th) Ave, Columbus, OH, USA
| | - Stephen Y Kang
- Department of Otolaryngology - Head and Neck Surgery, Ohio State University Wexner Medical Center, 410 W. 10(th) Ave, Columbus, OH, USA
| | - Sidharth V Puram
- Department of Otolaryngology - Head and Neck Surgery, Washington University School of Medicine, 4921 Parkview Pl, St. Louis, MO 63110, USA; Department of Genetics, Washington University School of Medicine, 4921 Parkview Pl, St. Louis, MO 63110, USA.
| |
Collapse
|
7
|
Li MM, Puram SV, Silverman DA, Old MO, Rocco JW, Kang SY. Margin Analysis in Head and Neck Cancer: State of the Art and Future Directions. Ann Surg Oncol 2019; 26:4070-4080. [PMID: 31385128 PMCID: PMC7382965 DOI: 10.1245/s10434-019-07645-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Indexed: 01/28/2023]
Abstract
BACKGROUND The status of surgical margins is the most important prognosticator for patients undergoing surgical resection of head and neck squamous cell carcinoma (HNSCC). Despite this, analysis of surgical margins is fraught with inconsistencies, including the ways in which margins are sampled and interpreted. Fundamentally, even the definition what constitutes a "clear" (or negative) margin may vary between institutions, surgeons, and pathologists. METHODS The PubMed database was queried for articles relevant to the topic, and experts in the field were consulted regarding key articles for inclusion. Abstracts were reviewed and the full text was accessed for articles of particular interest. RESULTS Data regarding various approaches to traditional margin analysis have been published without consensus. Several next-generation technologies have emerged in recent years that hold promise. CONCLUSION An overview and appraisal of traditional margin analysis techniques are provided. Additionally, we explore novel technologies that may assist in more accurate margin assessment, guide the extent of surgical resections intraoperatively, and inform decisions regarding adjuvant treatment postoperatively.
Collapse
Affiliation(s)
- Michael M Li
- Division of Head and Neck Oncology, Department of Otolaryngology - Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Sidharth V Puram
- Division of Head and Neck Oncology, Department of Otolaryngology - Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Dustin A Silverman
- Division of Head and Neck Oncology, Department of Otolaryngology - Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Matthew O Old
- Division of Head and Neck Oncology, Department of Otolaryngology - Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - James W Rocco
- Division of Head and Neck Oncology, Department of Otolaryngology - Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA
| | - Stephen Y Kang
- Division of Head and Neck Oncology, Department of Otolaryngology - Head and Neck Surgery, The James Cancer Hospital and Solove Research Institute, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
8
|
Debie P, Hernot S. Emerging Fluorescent Molecular Tracers to Guide Intra-Operative Surgical Decision-Making. Front Pharmacol 2019; 10:510. [PMID: 31139085 PMCID: PMC6527780 DOI: 10.3389/fphar.2019.00510] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 04/24/2019] [Indexed: 12/26/2022] Open
Abstract
Fluorescence imaging is an emerging technology that can provide real-time information about the operating field during cancer surgery. Non-specific fluorescent agents, used for the assessment of blood flow and sentinel lymph node detection, have so far dominated this field. However, over the last decade, several clinical studies have demonstrated the great potential of targeted fluorescent tracers to visualize tumor lesions in a more specific way. This has led to an exponential growth in the development of novel molecular fluorescent contrast agents. In this review, the design of fluorescent molecular tracers will be discussed, with particular attention for agents and approaches that are of interest for clinical translation.
Collapse
Affiliation(s)
| | - Sophie Hernot
- Laboratory for in vivo Cellular and Molecular Imaging (ICMI-BEFY/MIMA), Vrije Universiteit Brussel, Brussels, Belgium
| |
Collapse
|
9
|
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
|
10
|
Targeting CD44v6 for fluorescence-guided surgery in head and neck squamous cell carcinoma. Sci Rep 2018; 8:10467. [PMID: 29992954 PMCID: PMC6041314 DOI: 10.1038/s41598-018-28059-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2017] [Accepted: 06/08/2018] [Indexed: 02/01/2023] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) is an often highly invasive tumor, infiltrating functionally important tissue areas. Achieving complete tumor resection and preserving functionally relevant tissue structures depends on precise identification of tumor-free resection margins during surgery. Fluorescence-guided surgery (FGS), by intraoperative detection of tumor cells using a fluorescent tracer, may guide surgical excision and identify tumor-positive resection margins. Using a literature survey on potential surface molecules followed by immunohistochemical validation, we identified CD44 variant 6 (CD44v6) as a constitutively expressed antigen in the invasion zone of HNSCC lesions. The monoclonal anti-CD44v6 antibody BIWA was labeled with both a near-infrared fluorescent dye (IRDye800CW) and a radioactive label (Indium-111) and dual-modality imaging was applied in a locally invasive tumor mouse model. BIWA accurately detected human HNSCC xenografts in mice with a tumor uptake of 54 ± 11% ID/g and invasion regions with an accuracy of 94%. When dissected under clinical-like conditions, tumor remnants approximately 0.7 mm in diameter consisting of a few thousand cells were identified by fluorescence imaging, resulting in reliable dissection of invasive microregions. These data indicate that CD44v6 is a suitable target for reliable near-infrared detection and FGS of invasive HNSCC lesions in vivo.
Collapse
|
11
|
uPAR-targeted optical near-infrared (NIR) fluorescence imaging and PET for image-guided surgery in head and neck cancer: proof-of-concept in orthotopic xenograft model. Oncotarget 2017; 8:15407-15419. [PMID: 28039488 PMCID: PMC5362495 DOI: 10.18632/oncotarget.14282] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 11/30/2016] [Indexed: 12/29/2022] Open
Abstract
Purpose Urokinase-like Plasminogen Activator Receptor (uPAR) is overexpressed in a variety of carcinoma types, and therefore represents an attractive imaging target. The aim of this study was to assess the feasibility of two uPAR-targeted probes for PET and fluorescence tumor imaging in a human xenograft tongue cancer model. Experimental design and results Tumor growth of tongue cancer was monitored by bioluminescence imaging (BLI) and MRI. Either ICG-Glu-Glu-AE105 (fluorescent agent) or 64Cu-DOTA-AE105 (PET agent) was injected systemically, and fluorescence imaging or PET/CT imaging was performed. Tissue was collected for micro-fluorescence imaging and histology. A clear fluorescent signal was detected in the primary tumor with a mean in vivo tumor-to-background ratio of 2.5. Real-time fluorescence-guided tumor resection was possible, and sub-millimeter tumor deposits could be localized. Histological analysis showed co-localization of the fluorescent signal, uPAR expression and tumor deposits. In addition, the feasibility of uPAR-guided robotic cancer surgery was demonstrated. Also, uPAR-PET imaging showed a clear and localized signal in the tongue tumors. Conclusions This study demonstrated the feasibility of combining two uPAR-targeted probes in a preclinical head and neck cancer model. The PET modality provided preoperative non-invasive tumor imaging and the optical modality allowed for real-time fluorescence-guided tumor detection and resection. Clinical translation of this platform seems promising.
Collapse
|
12
|
Boonstra MC, Van Driel PBAA, Keereweer S, Prevoo HAJM, Stammes MA, Baart VM, Löwik CWGM, Mazar AP, van de Velde CJH, Vahrmeijer AL, Sier CFM. Preclinical uPAR-targeted multimodal imaging of locoregional oral cancer. Oral Oncol 2017; 66:1-8. [PMID: 28249642 DOI: 10.1016/j.oraloncology.2016.12.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Accepted: 12/27/2016] [Indexed: 12/13/2022]
Abstract
OBJECTIVES Establishing adequate resection margins and lymphatic mapping are crucial for the prognosis of oral cancer patients. Novel targeted imaging modalities are needed, enabling pre- and intraoperative detection of tumour cells, in combination with improved post-surgical examination by the pathologist. The urokinase-receptor (uPAR) is overexpressed in head and neck cancer, where it is associated with tumour progression and metastasis. MATERIAL AND METHODS To determine suitability of uPAR for molecular imaging of oral cancer surgery, human head and neck tumours were sectioned and stained for uPAR to evaluate the expression pattern compared to normal mucosa. Furthermore, metastatic oral squamous carcinoma cell line OSC-19 was used for targeting uPAR in in vivo mouse models. Using anti-uPAR antibody ATN-658, equipped with a multimodal label, the in vivo specificity was investigated and the optimal dose and time-window were evaluated. RESULTS All human oral cancer tissues expressed uPAR in epithelial and stromal cells. Hybrid ATN-658 clearly visualized tongue tumours in mice using either NIRF or SPECT imaging. Mean fluorescent TBRs over time were 4.3±0.7 with the specific tracer versus 1.7±0.1 with a control antibody. A significant difference in TBRs could be seen between 1nmol (150μg) and 0.34nmol (50μg) dose groups (n=4, p<0.05). Co-expression between BLI, GFP and the NIR fluorescent signals were seen in the tongue tumour, whereas human cytokeratin staining confirmed presence of malignant cells in the positive cervical lymph nodes. CONCLUSION This study shows the applicability of an uPAR specific multimodal tracer in an oral cancer model, combining SPECT with intraoperative guidance.
Collapse
Affiliation(s)
- M C Boonstra
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - P B A A Van Driel
- Department of Radiology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - S Keereweer
- Department of Otorhinolaryngology, and Head & Neck Surgery, Erasmus Medical Centre, Rotterdam, Netherlands
| | - H A J M Prevoo
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - M A Stammes
- Department of Radiology, Leiden University Medical Centre, Leiden, Netherlands; Percuros BV, Enschede, Netherlands
| | - V M Baart
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - C W G M Löwik
- Department of Radiology, Erasmus Medical Centre, Rotterdam, Netherlands
| | - A P Mazar
- Monopar Therapeutics Inc, Northbrook, IL, United States
| | - C J H van de Velde
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - A L Vahrmeijer
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands
| | - C F M Sier
- Department of Surgery, Leiden University Medical Centre, Leiden, Netherlands; Antibodies for Research Applications BV, Gouda, Netherlands.
| |
Collapse
|
13
|
Xie B, Stammes MA, van Driel PBAA, Cruz LJ, Knol-Blankevoort VT, Löwik MAM, Mezzanotte L, Que I, Chan A, van den Wijngaard JPHM, Siebes M, Gottschalk S, Razansky D, Ntziachristos V, Keereweer S, Horobin RW, Hoehn M, Kaijzel EL, van Beek ER, Snoeks TJA, Löwik CWGM. Necrosis avid near infrared fluorescent cyanines for imaging cell death and their use to monitor therapeutic efficacy in mouse tumor models. Oncotarget 2016; 6:39036-49. [PMID: 26472022 PMCID: PMC4770755 DOI: 10.18632/oncotarget.5498] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2015] [Accepted: 09/30/2015] [Indexed: 01/25/2023] Open
Abstract
Quantification of tumor necrosis in cancer patients is of diagnostic value as the amount of necrosis is correlated with disease prognosis and it could also be used to predict early efficacy of anti-cancer treatments. In the present study, we identified two near infrared fluorescent (NIRF) carboxylated cyanines, HQ5 and IRDye 800CW (800CW), which possess strong necrosis avidity. In vitro studies showed that both dyes selectively bind to cytoplasmic proteins of dead cells that have lost membrane integrity. Affinity for cytoplasmic proteins was confirmed using quantitative structure activity relations modeling. In vivo results, using NIRF and optoacoustic imaging, confirmed the necrosis avid properties of HQ5 and 800CW in a mouse 4T1 breast cancer tumor model of spontaneous necrosis. Finally, in a mouse EL4 lymphoma tumor model, already 24 h post chemotherapy, a significant increase in 800CW fluorescence intensity was observed in treated compared to untreated tumors. In conclusion, we show, for the first time, that the NIRF carboxylated cyanines HQ5 and 800CW possess strong necrosis avid properties in vitro and in vivo. When translated to the clinic, these dyes may be used for diagnostic or prognostic purposes and for monitoring in vivo tumor response early after the start of treatment.
Collapse
Affiliation(s)
- Bangwen Xie
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Marieke A Stammes
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands.,In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Pieter B A A van Driel
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands
| | - Luis J Cruz
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Vicky T Knol-Blankevoort
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands
| | - Martijn A M Löwik
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Laura Mezzanotte
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ivo Que
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Alan Chan
- Percuros BV, Enschede, The Netherlands
| | - Jeroen P H M van den Wijngaard
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Maria Siebes
- Department of Biomedical Engineering and Physics, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Sven Gottschalk
- Faculty of Medicine, Technical University of Munich, Munich, Germany.,Institute for Biological and Medical Imaging, Helmholtz Center Munich, Munich, Germany
| | - Daniel Razansky
- Faculty of Medicine, Technical University of Munich, Munich, Germany.,Institute for Biological and Medical Imaging, Helmholtz Center Munich, Munich, Germany
| | - Vasilis Ntziachristos
- Faculty of Medicine, Technical University of Munich, Munich, Germany.,Institute for Biological and Medical Imaging, Helmholtz Center Munich, Munich, Germany
| | - Stijn Keereweer
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Richard W Horobin
- School of Life Sciences, College of Medical, Veterinary and Life Sciences, The University of Glasgow, Glasgow, Scotland, UK
| | - Mathias Hoehn
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Percuros BV, Enschede, The Netherlands.,In-vivo-NMR Laboratory, Max Planck Institute for Neurological Research, Cologne, Germany
| | - Eric L Kaijzel
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Ermond R van Beek
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Medres, Cologne, Germany
| | - Thomas J A Snoeks
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Clemens W G M Löwik
- Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
14
|
Suganya S. AA, Kochurani KJ, Nair MG, Louis JM, Sankaran S, Rajagopal R, Kumar KS, Abraham P, P. G. B, Sebastian P, Somananthan T, Maliekal TT. TM1-IR680 peptide for assessment of surgical margin and lymph node metastasis in murine orthotopic model of oral cancer. Sci Rep 2016; 6:36726. [PMID: 27827443 PMCID: PMC5101486 DOI: 10.1038/srep36726] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Accepted: 10/20/2016] [Indexed: 11/08/2022] Open
Abstract
Treatment outcome after surgical removal in oral carcinoma is poor due to inadequate methodologies available for marking surgical margins. Even though some methodologies for intraoperative margin assessment are under clinical and preclinical trials for other solid tumours, a promising modality for oral cancer surgery is not developed. Fluorescent-based optical imaging using Near Infrared (NIR) dyes tagged to tumour specific target will be an optimal tool for this purpose. One such target, Gastrin Releasing Peptide Receptor (GRPR) was selected for the study, and its binding peptide, TM1-IR680, was tested for its efficacy for surgical margin prediction in murine orthotopic model of oral cancer, derived from primary samples. Here, for the first time in a preclinical analysis, we show that the size and margin of oral cancer can be predicted, as revealed by 3D-imaging. Interestingly, the peptide was sensitive enough to detect lymph nodes that harboured dispersed tumour cells before colonization, which was impossible to identify by conventional histopathology. We recommend the use of TM1-NIR dyes alone or in combination with other technologies to improve the clinical outcome of oral cancer surgery.
Collapse
Affiliation(s)
- Annie A. Suganya S.
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - K. J. Kochurani
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Madhumathy G. Nair
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Jiss Maria Louis
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Santhosh Sankaran
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - R. Rajagopal
- Animal Research Facility, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - K. Santhosh Kumar
- Chemical Biology Group, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Parvin Abraham
- Chemical Biology Group, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| | - Balagopal P. G.
- Surgical Oncology, Regional Cancer Centre, Thiruvananthapuram, Kerala, 695011, India
| | - Paul Sebastian
- Surgical Oncology, Regional Cancer Centre, Thiruvananthapuram, Kerala, 695011, India
| | - Thara Somananthan
- Division of Pathology, Regional Cancer Centre, Thiruvananthapuram, Kerala, 695011, India
| | - Tessy Thomas Maliekal
- Cancer Research Program, Rajiv Gandhi Centre for Biotechnology, Thiruvananthapuram, Kerala, 695014, India
| |
Collapse
|
15
|
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
|
16
|
Barroso EM, Smits RWH, van Lanschot CGF, Caspers PJ, Ten Hove I, Mast H, Sewnaik A, Hardillo JA, Meeuwis CA, Verdijk R, Noordhoek Hegt V, Baatenburg de Jong RJ, Wolvius EB, Bakker Schut TC, Koljenović S, Puppels GJ. Water Concentration Analysis by Raman Spectroscopy to Determine the Location of the Tumor Border in Oral Cancer Surgery. Cancer Res 2016; 76:5945-5953. [PMID: 27530325 DOI: 10.1158/0008-5472.can-16-1227] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2016] [Accepted: 07/06/2016] [Indexed: 11/16/2022]
Abstract
Adequate resection of oral cavity squamous cell carcinoma (OCSCC) means complete tumor removal with a clear margin of more than 5 mm. For OCSCC, 85% of the surgical resections appear inadequate. Raman spectroscopy is an objective and fast tool that can provide real-time information about the molecular composition of tissue and has the potential to provide an objective and fast intraoperative assessment of the entire resection surface. A previous study demonstrated that OCSCC can be discriminated from healthy surrounding tissue based on the higher water concentration in tumor. In this study, we investigated how the water concentration changes across the tumor border toward the healthy surrounding tissue on freshly excised specimens from the oral cavity. Experiments were performed on tissue sections from 20 patients undergoing surgery for OCSCC. A transition from a high to a lower water concentration, from tumor (76% ± 8% of water) toward healthy surrounding tissue (54% ± 24% of water), takes place over a distance of about 4 to 6 mm across the tumor border. This was accompanied by an increase of the heterogeneity of the water concentration in the surrounding healthy tissue. The water concentration distributions between the regions were significantly different (P < 0.0001). This new finding highlights the potential of Raman spectroscopy for objective intraoperative assessment of the resection margins. Cancer Res; 76(20); 5945-53. ©2016 AACR.
Collapse
Affiliation(s)
- Elisa M Barroso
- Department of Oral & Maxillofacial Surgery, Special Dental Care, and Orthodontics, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Roeland W H Smits
- Department of Otorhinolaryngology & Head and Neck Surgery, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Cornelia G F van Lanschot
- Department of Otorhinolaryngology & Head and Neck Surgery, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Peter J Caspers
- Center for Optical Diagnostics & Therapy, Department of Dermatology, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands. RiverD International BV, Rotterdam, The Netherlands
| | - Ivo Ten Hove
- Department of Oral & Maxillofacial Surgery, Special Dental Care, and Orthodontics, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Hetty Mast
- Department of Oral & Maxillofacial Surgery, Special Dental Care, and Orthodontics, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Aniel Sewnaik
- Department of Otorhinolaryngology & Head and Neck Surgery, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - José A Hardillo
- Department of Otorhinolaryngology & Head and Neck Surgery, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Cees A Meeuwis
- Department of Otorhinolaryngology & Head and Neck Surgery, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Rob Verdijk
- Department of Pathology, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | | | - Robert J Baatenburg de Jong
- Department of Otorhinolaryngology & Head and Neck Surgery, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Eppo B Wolvius
- Department of Oral & Maxillofacial Surgery, Special Dental Care, and Orthodontics, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Tom C Bakker Schut
- Center for Optical Diagnostics & Therapy, Department of Dermatology, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands. RiverD International BV, Rotterdam, The Netherlands.
| | - Senada Koljenović
- Department of Pathology, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands
| | - Gerwin J Puppels
- Center for Optical Diagnostics & Therapy, Department of Dermatology, Cancer Institute, Erasmus MC, Rotterdam, The Netherlands. RiverD International BV, Rotterdam, The Netherlands
| |
Collapse
|
17
|
Iqbal H, Pan Q. Image guided surgery in the management of head and neck cancer. Oral Oncol 2016; 57:32-9. [DOI: 10.1016/j.oraloncology.2016.04.007] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2015] [Revised: 04/11/2016] [Accepted: 04/12/2016] [Indexed: 12/15/2022]
|
18
|
de Boer E, Moore LS, Warram JM, Huang CC, Brandwein-Gensler MS, van Dam GM, Rosenthal EL, Schmalbach CE. On the horizon: Optical imaging for cutaneous squamous cell carcinoma. Head Neck 2015; 38 Suppl 1:E2204-13. [PMID: 25899874 DOI: 10.1002/hed.24079] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/13/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND Surgical resection with negative margins remains the standard of care for high-risk cutaneous squamous cell carcinoma (SCC). However, surgical management is often limited by poor intraoperative tumor visualization and inability to detect occult nodal metastasis. The inability to intraoperatively detect microscopic disease can lead to additional surgery, tumor recurrence, and decreased survival. METHODS A comprehensive literature review was conducted to identify studies incorporating optical imaging technology in the management of cutaneous SCC (January 1, 2000-December 1, 2014). RESULTS Several innovative optical imaging techniques, Raman spectroscopy, confocal microscopy, and fluorescence imaging, have been developed for intraoperative surgical guidance. Fifty-seven studies review the ability of these techniques to improve cutaneous SCC localization at the gross and microscopic level. CONCLUSION Significant advances have been achieved with real-time optical imaging strategies for intraoperative cutaneous SCC margin assessment and tumor detection. Optical imaging holds promise in improving the percentage of negative surgical margins and in the early detection of micrometastatic disease. © 2015 Wiley Periodicals, Inc. Head Neck 38: E2204-E2213, 2016.
Collapse
Affiliation(s)
- Esther de Boer
- Division of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama.,Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Lindsay S Moore
- Division of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Jason M Warram
- Division of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Conway C Huang
- Department of Dermatology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Margaret S Brandwein-Gensler
- Division of Anatomic Pathology, Department of Pathology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Gooitzen M van Dam
- Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Eben L Rosenthal
- Division of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Cecelia E Schmalbach
- Division of Otolaryngology, University of Alabama at Birmingham, Birmingham, Alabama
| |
Collapse
|
19
|
Kaur J, Jacobs R. Combination of Autofluorescence imaging and salivary protoporphyrin in Oral precancerous and cancerous lesions: Non-invasive tools. J Clin Exp Dent 2015; 7:e187-91. [PMID: 26155330 PMCID: PMC4483321 DOI: 10.4317/jced.52100] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2014] [Accepted: 11/27/2014] [Indexed: 11/07/2022] Open
Abstract
Background Normal and cancerous tissues have distinct auto-fluorescence properties because of differences in their biophysical and biochemical agents. Scientific evidences related to diagnostic fluorescence imaging for detection of oral precancerous and cancerous lesions are very limited. Objectives The aim of this study was to find out potential relationships between serum, salivary and tissue protoporphyrin IX ( PX) levels in subjects with or without oral precancerous and cancerous lesions. Also , to find out diagnostic value of fluorescence imaging (VELscope® system , LED Dental Inc., White Rock, B.C.) and salivary protoporphyrin IX (PX) in oral precancerous and cancerous lesions. Furthermore this study attempts to find out diagnostic value of the combination of approaches of fluorescence imaging and salivary protoporphyrin for detection of oral precancerous and cancerous lesions. Material and Methods The study sample comprised 3 test groups, with biopsy confirmed precancerous (leukoplakia and lichen planus) and cancerous lesions (squamous cell carcinoma) and one control group of 25 healthy individuals. To find out sensitivity and specificity, another 100 patients presenting for routine dental care were selected and clinical examinations were followed by fluorescence imaging and normal photography, which were finally confirmed by biopsy. The clinical and histopathogical examinations were done in conjunction with photography of the oral cavity using digital camera and fluorescence imaging. Serum, tissue and salivary protoporphyrin (PX) levels were measured. Results Using fluorescence imaging, oral cancerous and precancerous lesions showed deep purple to deep brown and dark green colour respectively, while normal tissues showed pale green colour in contrast. The PX levels in serum, salivary and tissues were significantly higher in precancerous and cancerous lesions as compared to normal healthy tissues. Salivary and serum PX levels were highly correlated in all groups. The sensitivity and specificity to the discrimination of precancerous and cancerous lesions from the healthy tissues were higher by combination approaches of salivary protoporphyrin X and VELscope® system as compared individual approach. Conclusions Combination approach of salivary protoporphyrin X and VELscope® system are more sensitive and specific to discriminate precancerous and cancerous lesions from the healthy tissues as compared to individual approach. Further studies are required on large samples of oral precancerous and cancerous lesions to test sensitivity and specificity and thus validate the clinical applicability of fluorescence imaging in (pre)cancerous diagnostics. Key words:Fluorescence imaging, oral cancerous, precancerous, protoporphyrin IX, saliva.
Collapse
Affiliation(s)
- Jasdeep Kaur
- BDS, MSc, OMFS IMPATH research group, Dept. Imaging & Pathology, Faculty of Medicine, University of Leuven and Oral & Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| | - Reinhilde Jacobs
- DDS, MSc, PhD, Dr h.c, OMFS IMPATH research group, Dept. Imaging & Pathology, Faculty of Medicine, University of Leuven and Oral & Maxillofacial Surgery, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
20
|
Karino M, Nakatani E, Hideshima K, Nariai Y, Tsunematsu K, Ohira K, Kanno T, Asahina I, Kagimura T, Sekine J. Applicability of preoperative nuclear morphometry to evaluating risk for cervical lymph node metastasis in oral squamous cell carcinoma. PLoS One 2014; 9:e116452. [PMID: 25549090 PMCID: PMC4280216 DOI: 10.1371/journal.pone.0116452] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 12/08/2014] [Indexed: 12/21/2022] Open
Abstract
BACKGROUND We previously reported the utility of preoperative nuclear morphometry for evaluating risk for cervical lymph node metastases in tongue squamous cell carcinoma. The risk for lymph node metastasis in oral squamous cell carcinoma, however, is known to differ depending on the anatomical site of the primary tumor, such as the tongue, gingiva, mouth floor, and buccal mucosa. In this study, we evaluated the applicability of this morphometric technique to evaluating the risk for cervical lymph node metastasis in oral squamous cell carcinoma. METHODS A digital image system was used to measure the mean nuclear area, mean nuclear perimeter, nuclear circular rate, ratio of nuclear length to width (aspect ratio), and nuclear area coefficient of variation (NACV). Relationships between these parameters and nodal status were evaluated by t-test and logistic regression analysis. RESULTS Eighty-eight cases of squamous cell carcinoma (52 of the tongue, 25 of the gingiva, 4 of the buccal mucosa, and 7 of the mouth floor) were included: 46 with positive node classification and 42 with negative node classification. Nuclear area and perimeter were significantly larger in node-positive cases than in node-negative cases; however, there were no significant differences in circular rate, aspect ratio, or NACV. We derived two risk models based on the results of multivariate analysis: Model 1, which identified age and mean nuclear area and Model 2, which identified age and mean nuclear perimeter. It should be noted that primary tumor site was not associated the pN-positive status. There were no significant differences in pathological nodal status by aspect ratio, NACV, or primary tumor site. CONCLUSION Our method of preoperative nuclear morphometry may contribute valuable information to evaluations of the risk for lymph node metastasis in oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Masaaki Karino
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Eiji Nakatani
- Translational Research Informatics Center, Kobe, Japan
| | - Katsumi Hideshima
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Yoshiki Nariai
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Kohji Tsunematsu
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Koichiro Ohira
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Takahiro Kanno
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
| | - Izumi Asahina
- Department of Regenerative Oral Surgery, Nagasaki University Graduate School of Medical Science, Nagasaki, Japan
| | | | - Joji Sekine
- Department of Oral and Maxillofacial Surgery, Shimane University Faculty of Medicine, Izumo, Japan
- * E-mail:
| |
Collapse
|
21
|
Conner KP, Rock BM, Kwon GK, Balthasar JP, Abuqayyas L, Wienkers LC, Rock DA. Evaluation of near infrared fluorescent labeling of monoclonal antibodies as a tool for tissue distribution. Drug Metab Dispos 2014; 42:1906-13. [PMID: 25209366 PMCID: PMC11024893 DOI: 10.1124/dmd.114.060319] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2014] [Accepted: 09/08/2014] [Indexed: 04/20/2024] Open
Abstract
The pharmacokinetic (PK) behavior of monoclonal antibodies (mAbs) is influenced by target-mediated drug disposition, off-target effects, antidrug antibody-mediated clearance, and interaction with fragment-crystallizable domain (Fc) receptors such as neonatal Fc receptor. All of these interactions hold the potential to impact mAb biodistribution. Near infrared (NIR) fluorescent probes offer an approach complementary to radionuclides to characterize drug disposition. Notably, the use of FDA-approved IRDye800 (IR800; LI-COR, Lincoln, NE) as a protein-labeling agent in preclinical work holds the potential for quantitative tissue analysis. Here, we tested the utility of the IR800 dye as a quantitative mAb tracer during pharmacokinetic analysis in both plasma and tissues using a model mouse monoclonal IgG1 (8C2) labeled with ≤1.5 molecules of IR800. The plasma PK parameters derived from a mixture of IR800-8C2 and 8C2 dosed intravenously to C57BL/6 mice at 8 mg/kg exhibited a large discrepancy in exposure depending on the method of quantitation [CLplasma = 8.4 ml/d per kilogram (NIR fluorescence detection) versus 2.5 ml/d per kilogram (enzyme-linked immunosorbent assay)]. The disagreement between measurements suggests that the PK of 8C2 is altered by addition of the IR800 dye. Additionally, direct fluorescence analysis of homogenized tissues revealed several large differences in IR800-8C2 tissue uptake when compared with a previously published study using [(125)I]8C2, most notably an over 4-fold increase in liver concentration. Finally, the utility of IR800 in combination with whole body imaging was examined by comparison of IR800-8C2 levels observed in animal sagittal cross-sections to those measured in homogenized tissues. Our results represent the first PK analysis in both mouse plasma and tissues of an IR800-mAb conjugate and suggest that mAb disposition is significantly altered by IR800 conjugation to 8C2.
Collapse
Affiliation(s)
- Kip P Conner
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Brooke M Rock
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Gayle K Kwon
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Joseph P Balthasar
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Lubna Abuqayyas
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Larry C Wienkers
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| | - Dan A Rock
- Biochemistry and Biophysics Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Seattle, Washington (K.P.C., B.M.R., G.K.K., L.C.W., D.A.R.); Department of Pharmaceutical Sciences, University at Buffalo, The State University of New York, Buffalo, New York (J.P.B.); and Quantitative Pharmacology Group in Pharmacokinetics and Drug Metabolism, Amgen Inc., Thousand Oaks, California (L.A.)
| |
Collapse
|
22
|
Atallah I, Milet C, Coll JL, Reyt E, Righini CA, Hurbin A. Role of near-infrared fluorescence imaging in head and neck cancer surgery: from animal models to humans. Eur Arch Otorhinolaryngol 2014; 272:2593-600. [PMID: 25115313 DOI: 10.1007/s00405-014-3224-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2014] [Accepted: 07/29/2014] [Indexed: 12/18/2022]
Abstract
Complete resection of head and neck cancers with negative surgical margins improves the prognosis of the disease and decreases the recurrence rate. Near-infrared fluorescence-guided surgery of head and neck cancer is a rapidly evolving field that represents an invaluable tool for tumor detection and resection. Here, we present a literature review of the principles of near-infrared fluorescence imaging and its use in head and neck cancer surgery. We discuss important studies in both animal models and humans that have been carried out up to this point. We also outline the important fluorescent molecules and devices used in head and neck fluorescence imaging-guided surgery. Although near-infrared fluorescence-guided surgery for head and neck cancers showed efficacy in animal models, its use in humans is limited by the small number of fluorescent probes that are approved for clinical use. However, it is considered as a novel surgical aid that helps delineate tumor margins preoperatively and could spare patients from the added morbidity that is associated with additional surgery or chemoradiation. In addition, it is a useful tool to detect sentinel lymph nodes as well as metastatic lymph nodes.
Collapse
Affiliation(s)
- Ihab Atallah
- Albert Bonniot Institute, CRI INSERM/UJF U823, BP170, 38042, Grenoble Cedex 9, France,
| | | | | | | | | | | |
Collapse
|
23
|
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
|
24
|
van Driel PBAA, van der Vorst JR, Verbeek FPR, Oliveira S, Snoeks TJA, Keereweer S, Chan B, Boonstra MC, Frangioni JV, van Bergen en Henegouwen PMP, Vahrmeijer AL, Lowik CWGM. Intraoperative fluorescence delineation of head and neck cancer with a fluorescent anti-epidermal growth factor receptor nanobody. Int J Cancer 2013; 134:2663-73. [PMID: 24222574 DOI: 10.1002/ijc.28601] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Accepted: 09/27/2013] [Indexed: 12/30/2022]
Abstract
Intraoperative near-infrared (NIR) fluorescence imaging is a technology with high potential to provide the surgeon with real-time visualization of tumors during surgery. Our study explores the feasibility for clinical translation of an epidermal growth factor receptor (EGFR)-targeting nanobody for intraoperative imaging and resection of orthotopic tongue tumors and cervical lymph node metastases. The anti-EGFR nanobody 7D12 and the negative control nanobody R2 were conjugated to the NIR fluorophore IRDye800CW (7D12-800CW and R2-800CW). Orthotopic tongue tumors were induced in nude mice using the OSC-19-luc2-cGFP cell line. Tumor-bearing mice were injected with 25 µg 7D12-800CW, R2-800CW or 11 µg 800CW. Subsequently, other mice were injected with 50 or 75 µg of 7D12-800CW. The FLARE imaging system and the IVIS spectrum were used to identify, delineate and resect the primary tumor and cervical lymph node metastases. All tumors could be clearly identified using 7D12-800CW. A significantly higher tumor-to-background ratio (TBR) was observed in mice injected with 7D12-800CW compared to mice injected with R2-800CW and 800CW. The highest average TBR (2.00 ± 0.34 and 2.72 ± 0.17 for FLARE and IVIS spectrum, respectively) was observed 24 hr after administration of the EGFR-specific nanobody. After injection of 75 µg 7D12-800CW cervical lymph node metastases could be clearly detected. Orthotopic tongue tumors and cervical lymph node metastases in a mouse model were clearly identified intraoperatively using a recently developed fluorescent EGFR-targeting nanobody. Translation of this approach to the clinic would potentially improve the rate of radical surgical resections.
Collapse
Affiliation(s)
- P B A A van Driel
- Department of Radiology and Molecular Imaging, Leiden University Medical Center, Leiden, The Netherlands; Percuros B.V., Enschede, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Grootendorst DJ, Steenbergen W, Manohar S, Ruers TJM. Optical techniques for the intraoperative assessment of nodal status. Future Oncol 2013; 9:1741-55. [PMID: 24156334 DOI: 10.2217/fon.13.125] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The lymphatic system is an important pathway in the metastatic spread of many malignancies and a key prognostic indicator. Nondestructive assessment of the nodal status during surgery could limit the amount of lymph nodes that need to be resected and allow for immediate regional lymphadenectomy during sentinel lymph node biopsy procedures. This review looks into the possibilities of conventional medical imaging methods that are capable of intraoperative nodal assessment and discusses multiple newly developed optical techniques. The physical background behind these techniques is reviewed and a concise overview of their main advantages and disadvantages is provided. These recent innovations show that while the application of optical modalities for intraoperative nodal staging is not yet applied routinely, there is reason enough to expect their introduction in the near future.
Collapse
Affiliation(s)
- Diederik J Grootendorst
- Biomedical Photonic Imaging Group, MIRA Institute for Biomedical Technology & Technical Medicine, Science & Technology, University of Twente, PO Box 217, 7500 AE Enschede, The Netherlands
| | | | | | | |
Collapse
|
27
|
Kobayashi H, Longmire MR, Choyke PL. Polychromatic in vivo imaging of multiple targets using visible and near infrared light. Adv Drug Deliv Rev 2013; 65:1112-9. [PMID: 23220327 DOI: 10.1016/j.addr.2012.10.015] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2012] [Revised: 10/25/2012] [Accepted: 10/31/2012] [Indexed: 12/01/2022]
Abstract
Conventional diagnostic imaging methods such as X-ray CT, MRI, and nuclear medicine are inherently monochromatic meaning that they can depict only one molecular target at a time. Optical imaging has the unique ability to be polychromatic and therefore multi-color imaging employing targeted agents conjugated to fluorophores of varying wavelength enables multiple simultaneous readouts thus providing greater multiplexed information. Numerous successful multicolor imaging techniques have recently been reported using optical imaging in in vivo animal disease models, thus adding to a growing body of research supporting the clinical viability and applicability of these technologies. Herein, we review multicolor optical imaging from the basic chemistry and physics perspective and then extend this to biological and medical applications.
Collapse
Affiliation(s)
- Hisataka Kobayashi
- Molecular Imaging Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, 10 Center Dr., Bethesda, MD 20892, USA.
| | | | | |
Collapse
|
28
|
Girardi FM, Zanella VG, Kroef RG. Correlation between clinical and pathological data and surgical margins in patients with squamous cell carcinoma of the oral cavity. Braz J Otorhinolaryngol 2013; 79:190-5. [PMID: 23670325 PMCID: PMC9443819 DOI: 10.5935/1808-8694.20130034] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2012] [Accepted: 12/29/2012] [Indexed: 11/20/2022] Open
Abstract
UNLABELLED The importance of having tumor-free margins when resecting oral neoplasms has been known for decades. OBJECTIVE To correlate clinical and pathology data to surgical margin status in patients with squamous cell carcinoma of the tongue and floor of the mouth. METHOD This historical cohort cross-sectional study included all patients submitted to squamous cell carcinoma resection for tumors of the oral tongue and floor of the mouth between 2007 and 2011 at the Head and Neck Surgery service of our institution. RESULTS In the 117 cases included, 68.3% had tongue tumors. The male-to-female ratio was 2.3:1 and patient mean age was 57.6 years. Broad free resection margins were seen in 23.0% of the cases; narrow margins in 60.6% of the cases; and compromised margins in 16.2%. Tumor diameter and thickness were correlated to resection margins. Tumors in more advanced T-stages presented more unsatisfactory margins. Patients operated with broad free margins had their tumors resected more commonly through transoral approaches. CONCLUSIONS Tumors of larger volume both in terms of diameter and thickness were more correlated to unsatisfactory resection margins. Higher complexity procedures were not associated with better resection margins.
Collapse
|
29
|
Kossatz S, Béhé M, Mansi R, Saur D, Czerney P, Kaiser WA, Hilger I. Multifactorial diagnostic NIR imaging of CCK2R expressing tumors. Biomaterials 2013; 34:5172-80. [PMID: 23591397 DOI: 10.1016/j.biomaterials.2013.03.073] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 03/23/2013] [Indexed: 11/28/2022]
Abstract
Optical imaging-based diagnostics identify malignancies based on molecular changes instead of morphological criteria in a non-invasive, irradiation free process. The aim of this study was to improve imaging efficiency by the development of a new Cholecystokinin-2-receptor targeted fluorescent peptide that matches the clinical needs regarding biodistribution and pharmacokinetics while displaying superior target specificity. Furthermore we performed multifactorial imaging of Cholecystokinin-2-receptor and tumor metabolism, since simultaneous targeting of various tumor biomarkers could intensely increase tumor identification and characterization. Affinity and specificity of the fluorescent Cholecystokinin-2-receptor targeted minigastrin (dQ-MG-754) were tested in vitro. We conducted in vivo imaging of the dQ-MG-754 probe alone and in a multifactorial approach with a GLUT-1 targeted probe (IR800 2-DG) on subcutaneous xenograft bearing athymic nude mice up to 24 h after intravenous injection (n = 5/group), followed by ex vivo biodistribution analysis and histological examination. We found specific, high affinity binding (Kd = 1.77 nM ± 0.6 nM) of dQ-MG-754 to Cholecystokinin-2-receptor expressing cells and xenografts as well as favorable pharmacokinetics for fluorescence-guided endoscopy. We successfully performed multifactorial imaging for the simultaneous detection of the Cholecystokinin-2-receptor and GLUT-1 targeted probe. Prominent differences in uptake patterns of the two contrast agents could be detected. The results were validated by histological examinations. The multifactorial imaging approach presented in this study could facilitate cancer detection in diagnostic imaging and intraoperative and endoscopic applications. Especially the dQ-MG-754 probe bears great potential for translation to clinical endoscopy imaging, because it combines specific high affinity binding with renal elimination and a favorable biodistribution.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Experimental Radiology, Institute of Diagnostic and Interventional Radiology I, Jena University Hospital, Friedrich Schiller University Jena, Jena, Germany.
| | | | | | | | | | | | | |
Collapse
|
30
|
Day KE, Beck LN, Heath CH, Huang CC, Zinn KR, Rosenthal EL. Identification of the optimal therapeutic antibody for fluorescent imaging of cutaneous squamous cell carcinoma. Cancer Biol Ther 2013; 14:271-7. [PMID: 23298904 DOI: 10.4161/cbt.23300] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
Intraoperative, real-time fluorescence imaging may significantly improve tumor visualization and resection and postoperatively, in pathological assessment. To this end, we sought to determine the optimal FDA approved therapeutic monoclonal antibody for optical imaging of human cutaneous squamous cell carcinoma (cSCC). A near-infrared (NIR) fluorescent probe (IRDye800) was covalently linked to bevacizumab, panitumumab or tocilizumab and injected systemically into immunodeficient mice bearing either cutaneous tumor cell lines (SCC13) or cutaneous human tumor explants. Tumors were then imaged and resected under fluorescent guidance with the SPY, an FDA-approved intraoperative imaging system, and the Pearl Impulse small animal imaging system. All fluorescently labeled antibodies delineated normal tissue from tumor in SCC13 xenografts based on tumor-to-background (TBR) ratios. The conjugated antibodies produced TBRs of 1.2-2 using SPY and 1.6-3.6 using Pearl; in comparison, isotype control antibody IgG-IRDye produced TBRs of 1.0 (SPY) and 0.98 (Pearl). Comparison between antibodies revealed them to be roughly equivalent for imaging purposes with both the SPY and Pearl (p = 0.89 SPY, p = 0.99 Pearl; one way ANOVA). Human tumor explants were also imaged and tumor detection was highest with panitumumab-IRDye800 when using the SPY (TBR 3.0) and Pearl (TBR 4.0). These data suggest that FDA approved antibodies may be clinically used for intraoperative detection of cSCC.
Collapse
Affiliation(s)
- Kristine E Day
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, USA
| | | | | | | | | | | |
Collapse
|
31
|
Thake M, Kalantzis A. Excision margins in squamous cell carcinoma of the tongue: A retrospective audit and review of the literature. ACTA ACUST UNITED AC 2013. [DOI: 10.4236/ojst.2013.31013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
32
|
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
|