1
|
Raman AP, Zachem TJ, Plumlee S, Park C, Eward W, Codd PJ, Ross W. Machine learning approaches in non-contact autofluorescence spectrum classification. PLOS DIGITAL HEALTH 2024; 3:e0000602. [PMID: 39383196 PMCID: PMC11463826 DOI: 10.1371/journal.pdig.0000602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Accepted: 06/10/2024] [Indexed: 10/11/2024]
Abstract
Manual surgical resection of soft tissue sarcoma tissue can involve many challenges, including the critical need for precise determination of tumor boundary with normal tissue and limitations of current surgical instrumentation, in addition to standard risks of infection or tissue healing difficulty. Substantial research has been conducted in the biomedical sensing landscape for development of non-human contact sensing devices. One such point-of-care platform, previously devised by our group, utilizes autofluorescence-based spectroscopic signatures to highlight important physiological differences in tumorous and healthy tissue. The following study builds on this work, implementing classification algorithms, including Artificial Neural Network, Support Vector Machine, Logistic Regression, and K-Nearest Neighbors, to diagnose freshly resected murine tissue as sarcoma or healthy. Classification accuracies of over 93% are achieved with Logistic Regression, and Area Under the Curve scores over 94% are achieved with Support Vector Machines, delineating a clear way to automate photonic diagnosis of ambiguous tissue in assistance of surgeons. These interpretable algorithms can also be linked to important physiological diagnostic indicators, unlike the black-box ANN architecture. This is the first known study to use machine learning to interpret data from a non-contact autofluorescence sensing device on sarcoma tissue, and has direct applications in rapid intraoperative sensing.
Collapse
Affiliation(s)
- Ashutosh P. Raman
- Department of Biomedical Engineering, Duke University, Durham, North Carolina, United States of America
| | - Tanner J. Zachem
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, United States of America
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Sarah Plumlee
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Christine Park
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - William Eward
- Department of Orthopaedic Surgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Patrick J. Codd
- Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina, United States of America
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| | - Weston Ross
- Department of Neurosurgery, Duke University School of Medicine, Durham, North Carolina, United States of America
| |
Collapse
|
2
|
Okay E, Gonzalez MR, Werenski JO, Sodhi A, Ozkan K, Brookes M, Ragbir M, Rankin K, Kumar AT, Lozano-Calderon SA. What is the diagnostic accuracy of fluorescence-guided surgery for margin assessment in appendicular bone and soft tissue tumors? - A systematic review of clinical studies. Surg Oncol 2024; 52:102030. [PMID: 38183855 DOI: 10.1016/j.suronc.2023.102030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/18/2023] [Accepted: 12/15/2023] [Indexed: 01/08/2024]
Abstract
BACKGROUND Fluorescence-guided surgery (FGS) is a novel technique to successfully assess surgical margins intraoperatively. Investigation and adoption of this technique in orthopaedic oncology remains limited. METHODS The PRISMA guidelines were followed for this manuscript. Our study was registered on PROSPERO (380520). Studies describing the use of FGS for resection of bone and soft tissue sarcomas (STS) on humans were included. Diagnostic performance metrics (sensitivity, specificity, positive predictive value [PPV], negative predictive value [NPV] and accuracy) and margin positivity rate were the outcomes assessed. RESULTS Critical appraisal using the Joanna Brigs Institute checklists showed significant concerns for study quality. Sensitivity of FGS ranged from 22.2 % to 100 % in three of the four studies assessing his metrics; one study in appendicular tumors in the pediatric population reported 0 % sensitivity in the three cases included. Specificity ranged from 9.38 % to 100 %. PPV ranged from 14.6 % to 70 % while NPV was between 53.3 % and 100 %. The diagnostic accuracy ranged from 21.62 % to 92.31 %. Margin positivity rate ranged from 2 % to 50 %, with six of the seven studies reporting values between 20 % and 50 %. CONCLUSIONS FSG is a feasible technique to assess tumor margins in bone and STS. Reported performance metrics and margin positivity rates vary widely between studies due to low study quality and high heterogeneity in dying protocols. LEVEL OF EVIDENCE Level III, diagnostic study.
Collapse
Affiliation(s)
- Erhan Okay
- Department of Orthopaedic Surgery, Istanbul Goztepe Prof. Dr.Suleyman Yalcin City Hospital, Medeniyet University, Istanbul, Turkey
| | - Marcos R Gonzalez
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Joseph O Werenski
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Alisha Sodhi
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Korhan Ozkan
- Department of Orthopaedic Surgery, Istanbul Goztepe Prof. Dr.Suleyman Yalcin City Hospital, Medeniyet University, Istanbul, Turkey
| | - Marcus Brookes
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Maniram Ragbir
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Kenneth Rankin
- North of England Bone and Soft Tissue Tumour Service, Royal Victoria Infirmary, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Anand Tn Kumar
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Santiago A Lozano-Calderon
- Division of Orthopaedic Oncology, Department of Orthopaedic Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
3
|
Bray J, Eward W, Breen M. Defining the relevance of surgical margins. Part two: Strategies to improve prediction of recurrence risk. Vet Comp Oncol 2023; 21:145-158. [PMID: 36745110 DOI: 10.1111/vco.12881] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 12/03/2022] [Accepted: 02/03/2023] [Indexed: 02/07/2023]
Abstract
Due to the complex nature of tumour biology and the integration between host tissues and molecular processes of the tumour cells, a continued reliance on the status of the microscopic cellular margin should not remain our only determinant of the success of a curative-intent surgery for patients with cancer. Based on current evidence, relying on a purely cellular focus to provide a binary indication of treatment success can provide an incomplete interpretation of potential outcome. A more holistic analysis of the cancer margin may be required. If we are to move ahead from our current situation - and allow treatment plans to be more intelligently tailored to meet the requirements of each individual tumour - we need to improve our utilisation of techniques that either improve recognition of residual tumour cells within the surgical field or enable a more comprehensive interrogation of tumour biology that identifies a risk of recurrence. In the second article in this series on defining the relevance of surgical margins, the authors discuss possible alternative strategies for margin assessment and evaluation in the canine and feline cancer patient. These strategies include considering adoption of the residual tumour classification scheme; intra-operative imaging systems including fluorescence-guided surgery, optical coherence tomography and Raman spectroscopy; molecular analysis and whole transcriptome analysis of tissues; and the development of a biologic index (nomogram). These techniques may allow evaluation of individual tumour biology and the status of the resection margin in ways that are different to our current techniques. Ultimately, these techniques seek to better define the risk of tumour recurrence following surgery and provide the surgeon and patient with more confidence in margin assessment.
Collapse
Affiliation(s)
| | - Will Eward
- Orthopedic Surgical Oncologist, Duke Cancer Center, Durham, North Carolina, USA
| | - Matthew Breen
- Oscar J. Fletcher Distinguished Professor of Comparative Oncology Genetics, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
4
|
Mickelson MA. Updated Concepts in Oncologic Surgery: Apocrine Gland Anal Sac Adenocarcinoma and Mast Cell Tumors. Vet Clin North Am Small Anim Pract 2022; 52:549-580. [DOI: 10.1016/j.cvsm.2021.12.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
5
|
Abstract
Cysteine cathepsins are proteases critical in physiopathological processes and show potential as targets or biomarkers for diseases and medical conditions. The 11 members of the cathepsin family are redundant in some cases but remarkably independent of others, demanding the development of both pan-cathepsin targeting tools as well as probes that are selective for specific cathepsins with little off-target activity. This review addresses the diverse design strategies that have been employed to accomplish this tailored selectivity among cysteine cathepsin targets and the imaging modalities incorporated. The power of these diverse tools is contextualized by briefly highlighting the nature of a few prominent cysteine cathepsins, their involvement in select diseases, and the application of cathepsin imaging probes in research spanning basic biochemical studies to clinical applications.
Collapse
Affiliation(s)
- Kelton A Schleyer
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| | - Lina Cui
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida, 1345 Center Dr, Gainesville, FL 32610, USA.
| |
Collapse
|
6
|
Ye Y, Sun WW, Xu RX, Selmic LE, Sun M. Intraoperative assessment of canine soft tissue sarcoma by deep learning enhanced optical coherence tomography. Vet Comp Oncol 2021; 19:624-631. [PMID: 34173314 DOI: 10.1111/vco.12747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 06/02/2021] [Accepted: 06/02/2021] [Indexed: 11/27/2022]
Abstract
Soft tissue sarcoma (STS) is a locally aggressive and infiltrative tumour in dogs. Surgical resection is the treatment of choice for local tumour control. Currently, post-operative pathology is performed for surgical margin assessment. Spectral-domain optical coherence tomography (OCT) has recently been evaluated for its value for surgical margin assessment in some tumour types in dogs. The purpose of this study was to develop an automatic diagnosis system that can assist clinicians in real-time for OCT image interpretation of tissues at surgical margins. We utilized a ResNet-50 network to classify healthy and cancerous tissues. A patch-based approach was adopted to achieve accurate classification with limited training data (80 cancer images, 80 normal images) and the validation set (20 cancer images, 20 normal images). The proposed method achieved an average accuracy of 97.1% with an excellent sensitivity of 94.3% on the validation set; the quadratic weighted κ was 0.94 for the STS diagnosis. In an independent test data set of 20 OCT images (10 cancer images, 10 normal images), the proposed method correctly differentiated all the STS images. Furthermore, we proposed a diagnostic curve, which could be evaluated in real-time to assist clinicians in detecting the specific location of a lesion. In short, the proposed method is accurate, operates in real-time and is non-invasive, which could be helpful for future surgical guidance.
Collapse
Affiliation(s)
- Yu Ye
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, China
| | - Weihong William Sun
- Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Ronald X Xu
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, China.,Department of Biomedical Engineering, The Ohio State University, Columbus, Ohio, USA
| | - Laura E Selmic
- Department of Veterinary Clinical Sciences, The Ohio State University, Columbus, Ohio, USA
| | - Mingzhai Sun
- Department of Precision Machinery and Precision Instrumentation, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
7
|
Dornbusch JA, Selmic LE, Huang PC, Samuelson JP, McLaughlin EM, Wavreille VA, Ogden JA, Abrams B, Kalamaras A, Green E, Hostnik ET, Every L, Fuerst JA, Jennings R, Premanandan C, Lorbach JN, Linn SC, Alex A, Sorrells JE, Yang L, Boppart SA. Diagnostic accuracy of optical coherence tomography for assessing surgical margins of canine soft tissue sarcomas in observers of different specialties. Vet Surg 2021; 50:111-120. [PMID: 32916007 PMCID: PMC9744383 DOI: 10.1111/vsu.13510] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Revised: 06/29/2020] [Accepted: 08/07/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE To determine the diagnostic accuracy of optical coherence tomography (OCT) to assess surgical margins of canine soft tissue sarcoma (STS) and determine the influence of observer specialty and training. STUDY DESIGN Blinded clinical prospective study. ANIMALS Twenty-five dogs undergoing surgical excision of STS. METHODS In vivo and ex vivo surgical margins were imaged with OCT after tumor resection. Representative images and videos were used to generate a training presentation and data sets. These were completed by 16 observers of four specialties (surgery, radiology, pathology, and OCT researchers). Images and videos from data sets were classified as cancerous or noncancerous. RESULTS The overall sensitivity and specificity were 88.2% and 92.8%, respectively, for in vivo tissues and 82.5% and 93.3%, respectively, for ex vivo specimens. The overall accurate classification for all specimens was 91.4% in vivo and 89.5% ex vivo. There was no difference in accuracy of interpretation of OCT imaging by observers of different specialties or experience levels. CONCLUSION Use of OCT to accurately assess surgical margins after STS excision was associated with a high sensitivity and specificity among various specialties. Personnel of all specialties and experience levels could effectively be trained to interpret OCT imaging. CLINICAL SIGNIFICANCE Optical coherence tomography can be used by personnel of different specialty experience levels and from various specialties to accurately identify canine STS in vivo and ex vivo after a short training session. These encouraging results provide evidence to justify further research to assess the ability of OCT to provide real-time assessments of surgical margins and its applicability to other neoplasms.
Collapse
Affiliation(s)
- Josephine A. Dornbusch
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Laura E. Selmic
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Pin-Chieh Huang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Jonathan P. Samuelson
- Department of Veterinary Clinical Medicine, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | | | - Vincent A. Wavreille
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Jessica A. Ogden
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Brittany Abrams
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Alex Kalamaras
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Eric Green
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Eric T. Hostnik
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Lincoln Every
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Jason A. Fuerst
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Ryan Jennings
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Christopher Premanandan
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Joshua N. Lorbach
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Sarah C. Linn
- Department of Veterinary Biosciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| | - Aneesh Alex
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Janet E. Sorrells
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Lingxiao Yang
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Stephen A. Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, Illinois,Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois,Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois
| |
Collapse
|
8
|
Abrams BE, Putterman AB, Ruple A, Wavreille V, Selmic LE. Variability in tumor margin reporting for soft tissue sarcoma and cutaneous mast cell tumors in dogs: A systematic review. Vet Surg 2020; 50:259-272. [DOI: 10.1111/vsu.13539] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2020] [Revised: 10/01/2020] [Accepted: 10/17/2020] [Indexed: 12/16/2022]
Affiliation(s)
- Brittany E. Abrams
- Department of Veterinary Clinical Sciences College of Veterinary Medicine, The Ohio State University Columbus Ohio
| | - Allison B. Putterman
- Department of Veterinary Clinical Medicine College of Veterinary Medicine, University of Illinois at Urbana‐Champaign Urbana Illinois
| | - Audrey Ruple
- Department of Comparative Pathobiology College of Veterinary Medicine, Purdue University West Lafayette, Indiana
| | - Vincent Wavreille
- Department of Veterinary Clinical Sciences College of Veterinary Medicine, The Ohio State University Columbus Ohio
| | - Laura E. Selmic
- Department of Veterinary Clinical Sciences College of Veterinary Medicine, The Ohio State University Columbus Ohio
| |
Collapse
|
9
|
Dornbusch JA, Cocca C, Jennings R, Samuelson J, Vieson M, Huang PC, Boppart SA, Wavreille VA, Selmic LE. The feasibility and utility of optical coherence tomography directed histopathology for surgical margin assessment of canine mast cell tumours. Vet Comp Oncol 2020; 19:616-623. [PMID: 32951309 DOI: 10.1111/vco.12654] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2020] [Revised: 09/17/2020] [Accepted: 09/17/2020] [Indexed: 12/18/2022]
Abstract
Histopathologic surgical margin assessment in veterinary patients is an imprecise science with assessment limited to a small proportion of the surgical margin due to time and finances. Incomplete excision of canine mast cell tumours (MCTs) alters treatment recommendations and prognosis. Optical coherence tomography (OCT) is a novel imaging modality that has been reported in a single veterinary study for surgical margin assessment. Twenty-five dogs with 34 MCTs were enrolled in a prospective pilot-study to assess the imaging characteristics of canine MCTs with OCT and to evaluate the feasibility and utility of OCT-guided histopathology. All dogs underwent routine surgical excision of MCTs. OCT imaging was used to assess the entire surgical margin prior to placement in formalin. Either normal areas or areas suspected of incomplete MCT excision were inked. Standard histopathologic sectioning and tangential sectioning of inked areas were performed and compared to OCT results. OCT identified MCT near the surgical margin in 10 of 26 specimens (38.4%). Four specimens suspicious for incomplete margins on OCT had incomplete MCT excision that was missed on standard histopathologic sectioning. Six specimens had OCT-guided sections taken as suspicious, which did not show MCT on histopathology. OCT-guided pathology sections were able to detect incompletely excised MCT near the surgical margin with a sensitivity of 90% and specificity of 56.2% in this preliminary study. OCT imaging shows promise for guiding pathologists to areas of interest to improve the diagnostic accuracy of surgical margin assessment in excised canine MCTs.
Collapse
Affiliation(s)
- Josephine A Dornbusch
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Ohio State University, Columbus, Ohio, USA
| | - Christina Cocca
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, USA
| | - Ryan Jennings
- Department of Veterinary Biosciences, College of Veterinary Medicine, Ohio State University, Columbus, Ohio, USA
| | - Jonathan Samuelson
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Miranda Vieson
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Pin-Chieh Huang
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana- Champaign, Urbana, Illinois, USA
| | - Stephen A Boppart
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana- Champaign, Urbana, Illinois, USA.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA.,Carle Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, USA
| | - Vincent A Wavreille
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Ohio State University, Columbus, Ohio, USA
| | - Laura E Selmic
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Ohio State University, Columbus, Ohio, USA
| |
Collapse
|
10
|
Favril S, Abma E, Stock E, Devriendt N, Van Goethem B, Blasi F, Brioschi C, Polis I, De Cock H, Miragoli L, Oliva P, Valbusa G, Vanderperren K, de Rooster H. Fluorescence-guided surgery using indocyanine green in dogs with superficial solid tumours. Vet Rec 2020; 187:273. [PMID: 32345608 DOI: 10.1136/vr.105554] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 04/01/2020] [Accepted: 04/05/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Near-infrared fluorescence (NIRF) imaging is a relatively novel technique that can aid surgeons during intraoperative tumour identification. METHODS Nine canine oncology patients (five mammary gland tumours, three mast cell tumours and one melanoma) received intravenous indocyanine green (ICG). After 24 hours, tumours were resected and fluorescence intensities of tumours and surroundings were evaluated. Additional wound bed tissue was resected if residual fluorescence was present after tumour resection. Ex vivo, fluorescence-guided dissection was performed to separate tumour from surrounding tissue. RESULTS Intraoperative NIRF-guided tumour delineation was feasible in four out of nine dogs. Wound bed imaging after tumour removal identified nine additional fluorescent lesions, of which four contained tumour tissue. One of these four true positive in vivo lesions was missed by standard-of-care inspection. Ex vivo fluorescence-guided tumour dissection showed a sensitivity of 72 per cent and a specificity of 80 per cent in discriminating between tumour and surrounding tissue. CONCLUSION The value of ICG for intraoperative tumour delineation seems more limited than originally thought. Although NIRF imaging using ICG did identify remaining tumour tissue in the wound bed, a high false positive rate was also observed.
Collapse
Affiliation(s)
- Sophie Favril
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium .,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| | - Emmelie Stock
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Nausikaa Devriendt
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Bart Van Goethem
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Ingeborgh Polis
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | | | | | - Paolo Oliva
- Bracco Imaging SpA, Colleretto Giacosa, Italy
| | | | - Katrien Vanderperren
- Department of Veterinary Medical Imaging and Small Animal Orthopaedics, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine, Ghent University, Merelbeke, Belgium.,Cancer Research Institute Ghent (CRIG), Medical Research Building, University Hospital, Ghent, Belgium
| |
Collapse
|
11
|
Holt D, Singhal S, Selmic LE. Near-infrared imaging and optical coherence tomography for intraoperative visualization of tumors. Vet Surg 2020; 49:33-43. [PMID: 31609011 PMCID: PMC11059208 DOI: 10.1111/vsu.13332] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2019] [Revised: 08/30/2019] [Accepted: 09/09/2019] [Indexed: 12/14/2022]
Abstract
Surgical excision is the foundation of treatment for early-stage solid tumors in man and companion animals. Complete excision with appropriate margins of surrounding tumor-free tissue is crucial to survival. Intraoperative imaging allows real-time visualization of tumors, assessment of surgical margins, and, potentially, lymph nodes and satellite metastatic lesions, allowing surgeons to perform complete tumor resections while sparing surrounding vital anatomic structures. This Review will focus on the use of near-infrared imaging and optical coherence tomography for intraoperative tumor visualization.
Collapse
Affiliation(s)
- David Holt
- Department of Clinical Sciences and Advanced Medicine, University of Pennsylvania School of Veterinary Medicine, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania
| | - Laura E Selmic
- Department of Veterinary Clinical Sciences, The Ohio State University College of Veterinary Medicine, Columbus, Ohio
| |
Collapse
|
12
|
Milovancev M, Tuohy JL, Townsend KL, Irvin VL. Influence of surgical margin completeness on risk of local tumour recurrence in canine cutaneous and subcutaneous soft tissue sarcoma: A systematic review and meta-analysis. Vet Comp Oncol 2019; 17:354-364. [PMID: 30953384 DOI: 10.1111/vco.12479] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 01/26/2019] [Accepted: 04/01/2019] [Indexed: 12/20/2022]
Abstract
The present peer-reviewed veterinary literature contains conflicting information regarding the impact of surgical margin completeness on risk of local tumour recurrence in canine soft tissue sarcoma (STS). This systematic review and meta-analysis was designed to answer the clinical question: "Does obtaining microscopically tumour-free surgical margins reduce risk for local tumour recurrence in canine cutaneous and subcutaneous STS?" A total of 486 citations were screened, 66 of which underwent full-text evaluation, with 10 studies representing 278 STS excisions ultimately included. Cumulatively, 16/164 (9.8%) of completely excised and 38/114 (33.3%) of incompletely excised STS recurred. Overall relative risk of 0.396 (95% confidence interval = 0.248-0.632) was calculated for local recurrence in STS excised with complete margins as compared to STS excised with incomplete margins. Risk of bias was judged to be low for all studies in terms of selection bias and detection bias but high for all studies in terms of performance bias and exclusion bias. The results of the present meta-analysis, coupled with the results of individual previous studies, strongly suggest that microscopically complete surgical margins confer a significantly reduced risk for local tumour recurrence in canine STS. Future studies ideally should adhere to standardized conducting and reporting guidelines to reduce systematic bias.
Collapse
Affiliation(s)
- Milan Milovancev
- Departments of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon
| | - Joanne L Tuohy
- Department of Clinical Sciences, College of Veterinary Medicine and Biomedical Sciences, Colorado State University, Fort Collins, Colorado
| | - Katy L Townsend
- Departments of Clinical Sciences, Carlson College of Veterinary Medicine, Oregon State University, Corvallis, Oregon
| | - Veronica L Irvin
- College of Public Health and Human Sciences, Oregon State University, Corvallis, Oregon
| |
Collapse
|
13
|
Selmic LE, Samuelson J, Reagan JK, Mesa KJ, Driskell E, Li J, Marjanovic M, Boppart SA. Intra-operative imaging of surgical margins of canine soft tissue sarcoma using optical coherence tomography. Vet Comp Oncol 2018; 17:80-88. [PMID: 30239117 DOI: 10.1111/vco.12448] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 09/12/2018] [Accepted: 09/13/2018] [Indexed: 01/15/2023]
Abstract
Optical coherence tomography (OCT) is a rapid non-invasive imaging technique that has shown high sensitivity for intra-operative surgical margin assessment in human breast cancer clinical trials. This promising technology has not been evaluated in veterinary medicine. The objective of this study was to correlate normal and abnormal histological features with OCT images for surgical margins from excised canine soft tissue sarcoma (STS) and to establish image evaluation criteria for identifying positive surgical margins. Fourteen client-owned dogs underwent surgical resection of a STS and OCT imaging of 2 to 4 areas of interest on the resected specimen were performed. Following imaging these areas were marked with surgical ink and trimmed for histopathology evaluation. Results showed that different tissue types had distinct characteristic appearances on OCT imaging. Adipose tissue exhibited a relatively low scattering and a honey-comb texture pattern. Skeletal muscle and sarcoma tissue were both dense and highly scattering. While sarcoma tissue was highly scattering, it did not have organized recognizable structure in contrast to muscle which showed clear fibre alignment patterns. In this investigation, we showed different tissue types had different and characteristic scattering and image texture appearances on OCT, which closely correlate with low-power histology images. Given the differentiation between tissue types the results support that OCT could be used to identify positive surgical margins immediately following resection of STS. Further research is needed to assess the diagnostic accuracy of this method for surgical margin assessment.
Collapse
Affiliation(s)
- Laura E Selmic
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Jonathan Samuelson
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Jennifer K Reagan
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Kelly J Mesa
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, Illinois.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Elizabeth Driskell
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Joanne Li
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, Illinois.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Marina Marjanovic
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, Illinois.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, Illinois
| | - Stephen A Boppart
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Champaign, Illinois.,Department of Electrical and Computer Engineering, University of Illinois at Urbana-Champaign, Champaign, Illinois.,Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, Illinois.,Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Champaign, Illinois
| |
Collapse
|
14
|
Favril S, Abma E, Blasi F, Stock E, Devriendt N, Vanderperren K, de Rooster H. Clinical use of organic near-infrared fluorescent contrast agents in image-guided oncologic procedures and its potential in veterinary oncology. Vet Rec 2018; 183:354. [DOI: 10.1136/vr.104851] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 04/11/2018] [Accepted: 04/17/2018] [Indexed: 12/15/2022]
Affiliation(s)
- Sophie Favril
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Eline Abma
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| | - Francesco Blasi
- Ephoran Multi-Imaging Solutions s.r.l.; Colleretto Giacosa Italy
| | - Emmelie Stock
- Department of Medical Imaging of Domestic Animals, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Nausikaa Devriendt
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Katrien Vanderperren
- Department of Medical Imaging of Domestic Animals, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
| | - Hilde de Rooster
- Small Animal Department, Faculty of Veterinary Medicine; Ghent University; Merelbeke Belgium
- Cancer Research Institute Ghent (CRIG); Ghent Belgium
| |
Collapse
|
15
|
New Techniques for Diagnosis and Treatment of Musculoskeletal Tumors: Methods of Intraoperative Margin Detection. Tech Orthop 2018. [DOI: 10.1097/bto.0000000000000290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
16
|
Beer P, Pozzi A, Rohrer Bley C, Bacon N, Pfammatter NS, Venzin C. The role of sentinel lymph node mapping in small animal veterinary medicine: A comparison with current approaches in human medicine. Vet Comp Oncol 2017; 16:178-187. [DOI: 10.1111/vco.12372] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Accepted: 10/17/2017] [Indexed: 12/20/2022]
Affiliation(s)
- P. Beer
- Clinic for Small Animal Surgery, Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| | - A. Pozzi
- Clinic for Small Animal Surgery, Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| | - C. Rohrer Bley
- Division of Radiation Oncology, Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| | - N. Bacon
- Fitzpatrick Referrals Oncology and Soft Tissue; Guildford Hospital; Guildford UK
| | - N. S. Pfammatter
- Clinic for Diagnostic Imaging, Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| | - C. Venzin
- Clinic for Small Animal Surgery, Vetsuisse Faculty; University of Zurich; Zurich Switzerland
| |
Collapse
|
17
|
Kramer L, Turk D, Turk B. The Future of Cysteine Cathepsins in Disease Management. Trends Pharmacol Sci 2017; 38:873-898. [PMID: 28668224 DOI: 10.1016/j.tips.2017.06.003] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 05/23/2017] [Accepted: 06/05/2017] [Indexed: 02/06/2023]
Abstract
Since the discovery of the key role of cathepsin K in bone resorption, cysteine cathepsins have been investigated by pharmaceutical companies as drug targets. The first clinical results from targeting cathepsins by activity-based probes and substrates are paving the way for the next generation of molecular diagnostic imaging, whereas the majority of antibody-drug conjugates currently in clinical trials depend on activation by cathepsins. Finally, cathepsins have emerged as suitable vehicles for targeted drug delivery. It is therefore timely to review the future of cathepsins in drug discovery. We focus here on inflammation-associated diseases because dysregulation of the immune system accompanied by elevated cathepsin activity is a common feature of these conditions.
Collapse
Affiliation(s)
- Lovro Kramer
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; International Postgraduate School Jozef Stefan, Jamova 39, 1000 Ljubljana, Slovenia
| | - Dušan Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKEBIP, Jamova 39, 1000 Ljubljana, Slovenia
| | - Boris Turk
- Jozef Stefan Institute, Department of Biochemistry and Molecular and Structural Biology, Jamova 39, 1000 Ljubljana, Slovenia; Center of Excellence CIPKEBIP, Jamova 39, 1000 Ljubljana, Slovenia; Faculty of Chemistry and Chemical Technology, University of Ljubljana, Večna pot 113, 1000 Ljubljana, Slovenia.
| |
Collapse
|
18
|
Milovancev M, Townsend KL, Gorman E, Bracha S, Curran K, Russell DS. Shaved margin histopathology and imprint cytology for assessment of excision in canine mast cell tumors and soft tissue sarcomas. Vet Surg 2017; 46:879-885. [DOI: 10.1111/vsu.12668] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2016] [Revised: 02/07/2017] [Accepted: 02/25/2017] [Indexed: 12/20/2022]
Affiliation(s)
- Milan Milovancev
- Department of Clinical Sciences, College of Veterinary Medicine; Oregon State University; Corvallis Oregon
| | - Kaitlin L. Townsend
- Department of Clinical Sciences, College of Veterinary Medicine; Oregon State University; Corvallis Oregon
| | - Elena Gorman
- Department of Biomedical Sciences, College of Veterinary Medicine; Oregon State University; Corvallis Oregon
| | - Shay Bracha
- Department of Clinical Sciences, College of Veterinary Medicine; Oregon State University; Corvallis Oregon
| | - Katie Curran
- Department of Clinical Sciences, College of Veterinary Medicine; Oregon State University; Corvallis Oregon
| | - Duncan S. Russell
- Department of Biomedical Sciences, College of Veterinary Medicine; Oregon State University; Corvallis Oregon
| |
Collapse
|
19
|
Milovancev M, Russell DS. Surgical margins in the veterinary cancer patient. Vet Comp Oncol 2017; 15:1136-1157. [PMID: 28194921 DOI: 10.1111/vco.12284] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 09/26/2016] [Accepted: 09/27/2016] [Indexed: 12/14/2022]
Abstract
In veterinary oncologic specimens, histopathology is the gold standard for determining adequacy of excision. Despite limitations of this technique, the pathologist's interpretation of margin status significantly impacts patient management, including indications for adjuvant therapy. This article aims to summarize peer-reviewed literature as it relates to histologic margin evaluation in veterinary cancer patients. The value of histologic tumour-free margins and technical factors influencing histopathologic margin outcomes are also discussed. We review alternative strategies for determining excisional status, and discuss how an evolving understanding of tumour biology might inform clinical and research perspectives on surgical margins. In doing so, we aim to provide context and a stimulus for future investigations into this important yet incompletely understood topic.
Collapse
Affiliation(s)
- M Milovancev
- Department of Veterinary Clinical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| | - D S Russell
- Department of Biomedical Sciences, Oregon State University, Corvallis, OR, 97331, USA
| |
Collapse
|
20
|
Bray JP. Soft tissue sarcoma in the dog - Part 2: surgical margins, controversies and a comparative review. J Small Anim Pract 2017; 58:63-72. [DOI: 10.1111/jsap.12629] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 07/07/2016] [Accepted: 08/08/2016] [Indexed: 12/12/2022]
Affiliation(s)
- J. P. Bray
- Companion Animal Clinical Studies, Head of Companion Animal Group, IVABS; Massey University; Palmerston North 4442 New Zealand
| |
Collapse
|
21
|
Chan CHF, Liesenfeld LF, Ferreiro-Neira I, Cusack JC. Preclinical Evaluation of Cathepsin-Based Fluorescent Imaging System for Cytoreductive Surgery. Ann Surg Oncol 2016; 24:931-938. [PMID: 27913947 DOI: 10.1245/s10434-016-5690-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2016] [Indexed: 12/21/2022]
Abstract
BACKGROUND Cytoreductive surgery and hyperthermic intraperitoneal chemotherapy (CRS-HIPEC) is a treatment option for peritoneal surface malignancies. The ability to detect microscopic foci of peritoneal metastasis intraoperatively may ensure the completeness of cytoreduction. In this study, we evaluated the suitability of a hand-held cathepsin-based fluorescent imaging system for intraoperative detection of appendiceal and colorectal peritoneal metastasis. METHODS Peritoneal tumors and normal peritoneal tissues were collected from patients with appendiceal and colorectal peritoneal metastasis. Expression of different cathepsins (CTS-B, -D, -F, -G, -K, -L, -O, and -S) was determined by quantitative RT-PCR and immunohistochemistry. The hand-held cathepsin-based fluorescent imaging system was used to detect peritoneal xenografts derived from human colon cancer cells (HT29, LoVo and HCT116) in nu/nu mice. RESULTS While the expression levels of CTS-B, -D, -L, and -S could be higher in peritoneal tumors than normal peritoneum with a median (range) of 6.1 (2.9-25.8), 2.0 (1.0-15.8), 1.4 (0.8-7.0), and 2.1 (1.6-13.9) folds by quantitative RT-PCR, respectively, CTS-B was consistently the major contributor of the overall cathepsin expression in appendiceal and colonic peritoneal tumors, including adenocarcinomas and low-grade appendiceal mucinous neoplasms. Using peritoneal xenograft mouse models, small barely visible colonic peritoneal tumors (<2.5 mm in maximum diameter) could be detected by the hand-held cathepsin-based fluorescent imaging system. CONCLUSIONS Because cathepsin expression is higher in peritoneal tumors than underlying peritoneum, the hand-held cathepsin-based fluorescent imaging system could be useful for intraoperative detection of microscopic peritoneal metastasis during CRS-HIPEC and clinical trial is warranted.
Collapse
Affiliation(s)
- Carlos H F Chan
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA. .,Department of Surgery, University of Iowa Carver College of Medicine, Iowa City, IA, USA.
| | | | | | - James C Cusack
- Department of Surgery, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
22
|
Nguyen JQ, Gowani ZS, O'Connor M, Pence IJ, Nguyen TQ, Holt GE, Schwartz HS, Halpern JL, Mahadevan-Jansen A. Intraoperative Raman spectroscopy of soft tissue sarcomas. Lasers Surg Med 2016; 48:774-781. [PMID: 27454580 DOI: 10.1002/lsm.22564] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2016] [Indexed: 01/21/2023]
Abstract
BACKGROUND AND OBJECTIVE Soft tissue sarcomas (STS) are a rare and heterogeneous group of malignant tumors that are often treated through surgical resection. Current intraoperative margin assessment methods are limited and highlight the need for an improved approach with respect to time and specificity. Here we investigate the potential of near-infrared Raman spectroscopy for the intraoperative differentiation of STS from surrounding normal tissue. MATERIALS AND METHODS In vivo Raman measurements at 785 nm excitation were intraoperatively acquired from subjects undergoing STS resection using a probe based spectroscopy system. A multivariate classification algorithm was developed in order to automatically identify spectral features that can be used to differentiate STS from the surrounding normal muscle and fat. The classification algorithm was subsequently tested using leave-one-subject-out cross-validation. RESULTS With the exclusion of well-differentiated liposarcomas, the algorithm was able to classify STS from the surrounding normal muscle and fat with a sensitivity and specificity of 89.5% and 96.4%, respectively. CONCLUSION These results suggest that single point near-infrared Raman spectroscopy could be utilized as a rapid and non-destructive surgical guidance tool for identifying abnormal tissue margins in need of further excision. Lasers Surg. Med. 48:774-781, 2016. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- John Q Nguyen
- Biophotonics Center, Vanderbilt University, 410 24th Ave. South (Keck FEL Center), Nashville, Tennessee 37232
| | - Zain S Gowani
- School of Medicine, Vanderbilt University, 2215 Garland Ave (Light Hall), Nashville, Tennessee 37232
| | - Maggie O'Connor
- Biophotonics Center, Vanderbilt University, 410 24th Ave. South (Keck FEL Center), Nashville, Tennessee 37232
| | - Isaac J Pence
- Biophotonics Center, Vanderbilt University, 410 24th Ave. South (Keck FEL Center), Nashville, Tennessee 37232
| | - The-Quyen Nguyen
- Department of Biomedical Engineering, Northwestern University, Silverman Hall, Evanston, Illinois 60208
| | - Ginger E Holt
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 691 Preston Building, Nashville, Tennessee 37232-6838
| | - Herbert S Schwartz
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 691 Preston Building, Nashville, Tennessee 37232-6838
| | - Jennifer L Halpern
- Vanderbilt-Ingram Cancer Center, Vanderbilt University Medical Center, 691 Preston Building, Nashville, Tennessee 37232-6838
| | - Anita Mahadevan-Jansen
- Biophotonics Center, Vanderbilt University, 410 24th Ave. South (Keck FEL Center), Nashville, Tennessee 37232.
| |
Collapse
|
23
|
Bartholf DeWitt S, Eward WC, Eward CA, Lazarides AL, Whitley MJ, Ferrer JM, Brigman BE, Kirsch DG, Berg J. A Novel Imaging System Distinguishes Neoplastic from Normal Tissue During Resection of Soft Tissue Sarcomas and Mast Cell Tumors in Dogs. Vet Surg 2016; 45:715-22. [PMID: 27281113 DOI: 10.1111/vsu.12487] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
OBJECTIVE To assess the ability of a novel imaging system designed for intraoperative detection of residual cancer in tumor beds to distinguish neoplastic from normal tissue in dogs undergoing resection of soft tissue sarcoma (STS) and mast cell tumor (MCT). STUDY DESIGN Non-randomized prospective clinical trial. ANIMALS 12 dogs with STS and 7 dogs with MCT. METHODS A fluorescent imaging agent that is activated by proteases in vivo was administered to the dogs 4-6 or 24-26 hours before tumor resection. During surgery, a handheld imaging device was used to measure fluorescence intensity within the cancerous portion of the resected specimen and determine an intensity threshold for subsequent identification of cancer. Selected areas within the resected specimen and tumor bed were then imaged, and biopsies (n=101) were obtained from areas that did or did not have a fluorescence intensity exceeding the threshold. Results of intraoperative fluorescence and histology were compared. RESULTS The imaging system correctly distinguished cancer from normal tissue in 93/101 biopsies (92%). Using histology as the reference, the sensitivity and specificity of the imaging system for identification of cancer in biopsies were 92% and 92%, respectively. There were 10/19 (53%) dogs which exhibited transient facial erythema soon after injection of the imaging agent which responded to but was not consistently prevented by intravenous diphenhydramine. CONCLUSION A fluorescence-based imaging system designed for intraoperative use can distinguish canine soft tissue sarcoma (STS) and mast cell tumor (MCT) tissue from normal tissue with a high degree of accuracy. The system has potential to assist surgeons in assessing the adequacy of tumor resections during surgery, potentially reducing the risk of local tumor recurrence. Although responsive to antihistamines, the risk of hypersensitivity needs to be considered in light of the potential benefits of this imaging system in dogs.
Collapse
Affiliation(s)
| | | | - Cindy A Eward
- Veterinary Specialty Hospital of the Carolinas, Cary, North Carolina
| | | | | | | | | | | | - John Berg
- Cummings School of Veterinary Medicine, Tufts University, North Grafton, Massachusetts
| |
Collapse
|
24
|
Pulz LH, Strefezzi RF. Proteases as prognostic markers in human and canine cancers. Vet Comp Oncol 2016; 15:669-683. [PMID: 27136601 DOI: 10.1111/vco.12223] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 02/07/2016] [Indexed: 02/05/2023]
Abstract
The extracellular matrix (ECM) is composed of several types of proteins, which interact and form dynamic networks. These components can modulate cell behaviour and actively influence the growth and differentiation of tissues. ECM is also important in several pathological processes, such as cancer invasion and metastasis, by creating favourable microenvironments. Proteolysis in neoplastic tissues is mediated by proteinases, whose regulation involves complex interactions between neoplastic cells and non-neoplastic stromal cells. In this review, we discuss aspects of proteinase expression and tumor behaviour in humans and dogs. Different classes of proteases are summarized, with special emphasis being placed on molecules that have been shown to correlate with prognosis, reinforcing the need for a better understanding of the regulation of this microenvironment and its influences in tumor progression and metastasis, which should significantly aid the development of improved prognosis and treatment.
Collapse
Affiliation(s)
- L H Pulz
- Laboratório de Oncologia Comparada e Translacional (LOCT), Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| | - R F Strefezzi
- Laboratório de Oncologia Comparada e Translacional (LOCT), Departamento de Medicina Veterinária, Faculdade de Zootecnia e Engenharia de Alimentos, Universidade de São Paulo, Pirassununga, Brazil
| |
Collapse
|
25
|
Cabon Q, Sayag D, Texier I, Navarro F, Boisgard R, Virieux-Watrelot D, Ponce F, Carozzo C. Evaluation of intraoperative fluorescence imaging-guided surgery in cancer-bearing dogs: a prospective proof-of-concept phase II study in 9 cases. Transl Res 2016; 170:73-88. [PMID: 26746803 DOI: 10.1016/j.trsl.2015.12.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2015] [Revised: 12/04/2015] [Accepted: 12/09/2015] [Indexed: 01/26/2023]
Abstract
The objective was to prospectively evaluate the application of intraoperative fluorescence imaging (IOFI) in the surgical excision of malignant masses in dogs, using a novel lipid nanoparticle contrast agent. Dogs presenting with spontaneous soft-tissue sarcoma or subcutaneous tumors were prospectively enrolled. Clinical staging and whole-body computed tomography (CT) were performed. All the dogs received an intravenous injection of dye-loaded lipid nanoparticles, LipImage 815. Wide or radical resection was realized after CT examination. Real-time IOFI was performed before skin incision and after tumor excision. In cases of radical resection, the lymph nodes (LNs) were imaged. The margin/healthy tissues fluorescence ratio or LN/healthy tissues fluorescence ratio was measured and compared with the histologic margins or LN status. Nine dogs were included. Limb amputation was performed in 3 dogs, and wide resection in 6. No adverse effect was noted. Fluorescence was observed in all 9 of the tumors. The margins were clean in 5 of 6 dogs after wide surgical resection, and the margin/healthy tissues fluorescence ratio was close to 1.0 in all these dogs. Infiltrated margins were observed in 1 case, with a margin/healthy tissues fluorescence ratio of 3.2. Metastasis was confirmed in 2 of 3 LNs, associated with LN/healthy tissues fluorescence ratios of 2.1 and 4.2, whereas nonmetastatic LN was associated with a ratio of 1.0. LipImage 815 used as a contrast agent during IOFI seemed to allow for good discrimination between tumoral and healthy tissues. Future studies are scheduled to evaluate the sensitivity and specificity of IOFI using LipImage 815 as a tracer.
Collapse
Affiliation(s)
- Quentin Cabon
- From the Surgery and Anesthesia Unit, VetAgro-Sup Campus Vétérinaire de Lyon, Marcy l'Etoile, France.
| | - David Sayag
- Clinical Oncology Department, Small Animal Internal Medicine Unit, VetAgro-Sup Campus Vétérinaire de Lyon, Marcy l'Etoile, France
| | - Isabelle Texier
- Université Grenoble Alpes, Grenoble, France; CEA-LETI MINATEC/ DTBS, Grenoble, France.
| | - Fabrice Navarro
- Université Grenoble Alpes, Grenoble, France; CEA-LETI MINATEC/ DTBS, Grenoble, France
| | | | | | - Frédérique Ponce
- Clinical Oncology Department, Small Animal Internal Medicine Unit, VetAgro-Sup Campus Vétérinaire de Lyon, Marcy l'Etoile, France
| | - Claude Carozzo
- From the Surgery and Anesthesia Unit, VetAgro-Sup Campus Vétérinaire de Lyon, Marcy l'Etoile, France
| |
Collapse
|
26
|
Sayag D, Cabon Q, Texier I, Navarro FP, Boisgard R, Virieux-Watrelot D, Carozzo C, Ponce F. Phase-0/phase-I study of dye-loaded lipid nanoparticles for near-infrared fluorescence imaging in healthy dogs. Eur J Pharm Biopharm 2016; 100:85-93. [PMID: 26777342 DOI: 10.1016/j.ejpb.2016.01.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2015] [Revised: 11/28/2015] [Accepted: 01/04/2016] [Indexed: 01/12/2023]
Abstract
Near-infrared (NIR) fluorescence imaging using FDA-approved indocyanine green (ICG) has been the subject of numerous studies during the past few years. It could constitute a potentially exciting new paradigm shift in veterinary oncology, especially to develop in vivo fluorescence imaging diagnostics and surgery guidance methods. The objective of this study was to evaluate the pharmacologic and toxicological characteristics in healthy beagle dogs of LipImage™ 815, a formulation made of NIR-dye-loaded lipid nanoparticles. The initial dosage for the evaluation of biodistribution was extrapolated from data in mice and then adapted to define the more adapted dose (MAD) according to the fluorescence results obtained in 5 dogs using a Fluobeam® 800 imaging device (phase 0 study). A single dose acute toxicity study was then performed (3 dogs, phase I study). Before the systemic administration of LipImage™ 815, the dogs presented a very mild residual fluorescence, particularly in the liver and kidneys. After injection, the plasma fluorescence continuously decreased, and the signal was relatively homogeneously distributed throughout the different organs, though more pronounced in the liver and to a lesser extent in the steroid-rich organs (adrenal, ovaries), intestines, lymph nodes and kidneys. A MAD of 2.0μg/kg was found. No evidence of acute or delayed general, hepatic, renal or hematologic toxicity was observed at 1-fold, 5-fold or 10-fold MAD. The results of this phase-0/phase-I study showed that an optimal dosage of LipImage™ 815 of 2.0μg/kg allowed the achievement of a fluorescence signal suitable for surgery guidance application without any acute side effects.
Collapse
Affiliation(s)
- David Sayag
- Clinical Oncology Department, Small Animal Internal Medicine Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, F-69280 Marcy l'Etoile, France.
| | - Quentin Cabon
- Surgery and Anesthesia Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, F-69280 Marcy l'Etoile, France
| | - Isabelle Texier
- Univ. Grenoble Alpes, F-38000 Grenoble, France; CEA LETI, MINATEC Campus, Technologies for Healthcare and Biology Division, 17 rue des Martyrs, F-38054 Grenoble, France.
| | - Fabrice P Navarro
- Univ. Grenoble Alpes, F-38000 Grenoble, France; CEA LETI, MINATEC Campus, Technologies for Healthcare and Biology Division, 17 rue des Martyrs, F-38054 Grenoble, France
| | - Raphaël Boisgard
- CEA I2BM SHFJ INSERM U1023, 4 place du Général, Leclerc, 91400 Orsay, France
| | - Dorothée Virieux-Watrelot
- Pathology Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, 69280 Marcy l'Etoile, France; ICE 2011-03-101 Research Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, 69280 Marcy l'Etoile, France
| | - Claude Carozzo
- Surgery and Anesthesia Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, F-69280 Marcy l'Etoile, France; ICE 2011-03-101 Research Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, 69280 Marcy l'Etoile, France
| | - Frédérique Ponce
- Clinical Oncology Department, Small Animal Internal Medicine Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, F-69280 Marcy l'Etoile, France; ICE 2011-03-101 Research Unit, VetAgro Sup Campus Vétérinaire de Lyon, 1 avenue Bourgelat, 69280 Marcy l'Etoile, France
| |
Collapse
|
27
|
Lazarides AL, Whitley MJ, Strasfeld DB, Cardona DM, Ferrer JM, Mueller JL, Fu HL, DeWitt SB, Brigman BE, Ramanujam N, Kirsch DG, Eward WC. A Fluorescence-Guided Laser Ablation System for Removal of Residual Cancer in a Mouse Model of Soft Tissue Sarcoma. Am J Cancer Res 2016; 6:155-66. [PMID: 26877775 PMCID: PMC4729765 DOI: 10.7150/thno.13536] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 09/28/2015] [Indexed: 12/22/2022] Open
Abstract
The treatment of soft tissue sarcoma (STS) generally involves tumor excision with a wide margin. Although advances in fluorescence imaging make real-time detection of cancer possible, removal is limited by the precision of the human eye and hand. Here, we describe a novel pulsed Nd:YAG laser ablation system that, when used in conjunction with a previously described molecular imaging system, can identify and ablate cancer in vivo. Mice with primary STS were injected with the protease-activatable probe LUM015 to label tumors. Resected tissues from the mice were then imaged and treated with the laser using the paired fluorescence-imaging/ laser ablation device, generating ablation clefts with sub-millimeter precision and minimal underlying tissue damage. Laser ablation was guided by fluorescence to target tumor tissues, avoiding normal structures. The selective ablation of tumor implants in vivo improved recurrence-free survival after tumor resection in a cohort of 14 mice compared to 12 mice that received no ablative therapy. This prototype system has the potential to be modified so that it can be used during surgery to improve recurrence-free survival in patients with cancer.
Collapse
|
28
|
Visgauss JD, Eward WC, Brigman BE. Innovations in Intraoperative Tumor Visualization. Orthop Clin North Am 2016; 47:253-64. [PMID: 26614939 DOI: 10.1016/j.ocl.2015.08.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
In the surgical management of solid tumors, adequacy of tumor resection has implications for local recurrence and survival. The standard method of intraoperative identification of tumor margin is frozen section pathologic analysis, which is time-consuming with potential for sampling error. Intraoperative tumor visualization has the potential to significantly improve surgical cancer care across disciplines, by guiding accuracy of biopsies, increasing adequacy of resections, directing adjuvant therapy, and even providing diagnostic information. We provide an outline of various methods of intraoperative tumor visualization developed to aid in the real-time assessment of tumor extent and adequacy of resection.
Collapse
Affiliation(s)
- Julia D Visgauss
- Department of Orthopaedic Surgery, Duke University, Box 3312 DUMC, Durham, NC 27710, USA
| | - William C Eward
- Department of Orthopaedic Surgery, Duke University, Box 3312 DUMC, Durham, NC 27710, USA
| | - Brian E Brigman
- Department of Orthopaedic Surgery, Duke University, Box 3312 DUMC, Durham, NC 27710, USA.
| |
Collapse
|
29
|
Mueller JL, Fu HL, Mito JK, Whitley MJ, Chitalia R, Erkanli A, Dodd L, Cardona DM, Geradts J, Willett RM, Kirsch DG, Ramanujam N. A quantitative microscopic approach to predict local recurrence based on in vivo intraoperative imaging of sarcoma tumor margins. Int J Cancer 2015; 137:2403-12. [PMID: 25994353 DOI: 10.1002/ijc.29611] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2015] [Accepted: 04/30/2015] [Indexed: 11/12/2022]
Abstract
The goal of resection of soft tissue sarcomas located in the extremity is to preserve limb function while completely excising the tumor with a margin of normal tissue. With surgery alone, one-third of patients with soft tissue sarcoma of the extremity will have local recurrence due to microscopic residual disease in the tumor bed. Currently, a limited number of intraoperative pathology-based techniques are used to assess margin status; however, few have been widely adopted due to sampling error and time constraints. To aid in intraoperative diagnosis, we developed a quantitative optical microscopy toolbox, which includes acriflavine staining, fluorescence microscopy, and analytic techniques called sparse component analysis and circle transform to yield quantitative diagnosis of tumor margins. A series of variables were quantified from images of resected primary sarcomas and used to optimize a multivariate model. The sensitivity and specificity for differentiating positive from negative ex vivo resected tumor margins was 82 and 75%. The utility of this approach was tested by imaging the in vivo tumor cavities from 34 mice after resection of a sarcoma with local recurrence as a bench mark. When applied prospectively to images from the tumor cavity, the sensitivity and specificity for differentiating local recurrence was 78 and 82%. For comparison, if pathology was used to predict local recurrence in this data set, it would achieve a sensitivity of 29% and a specificity of 71%. These results indicate a robust approach for detecting microscopic residual disease, which is an effective predictor of local recurrence.
Collapse
Affiliation(s)
- Jenna L Mueller
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Henry L Fu
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Jeffrey K Mito
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Melodi J Whitley
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina
| | - Rhea Chitalia
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| | - Alaattin Erkanli
- Department of Biostatistics & Bioinformatics, Duke University, Durham, North Carolina
| | - Leslie Dodd
- Department of Pathology, University of North Carolina School of Medicine, Chapel Hill, North Carolina
| | - Diana M Cardona
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Joseph Geradts
- Department of Pathology, Duke University Medical Center, Durham, North Carolina
| | - Rebecca M Willett
- Department of Electrical and Computer Engineering, University of Wisconsin-Madison, Madison, Wisconsin
| | - David G Kirsch
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, North Carolina.,Department of Radiation Oncology, Duke University School of Medicine, Durham, North Carolina
| | - Nimmi Ramanujam
- Department of Biomedical Engineering, Duke University, Durham, North Carolina
| |
Collapse
|
30
|
Rosenthal EL, Warram JM, Bland KI, Zinn KR. The status of contemporary image-guided modalities in oncologic surgery. Ann Surg 2015; 261:46-55. [PMID: 25599326 DOI: 10.1097/sla.0000000000000622] [Citation(s) in RCA: 95] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVE To review the current trends in optical imaging to guide oncologic surgery. BACKGROUND Surgical resection remains the cornerstone of therapy for patients with early stage solid malignancies and more than half of all patients with cancer undergo surgery each year. The technical ability of the surgeon to obtain clear surgical margins at the initial resection remains crucial to improve overall survival and long-term morbidity. Current resection techniques are largely based on subjective and subtle changes associated with tissue distortion by invasive cancer. As a result, positive surgical margins occur in a significant portion of tumor resections, which is directly correlated with a poor outcome. METHODS A comprehensive review of studies evaluating optical imaging techniques is performed. RESULTS A variety of cancer imaging techniques have been adapted or developed for intraoperative surgical guidance that have been shown to improve functional and oncologic outcomes in randomized clinical trials. There are also a large number of novel, cancer-specific contrast agents that are in early stage clinical trials and preclinical development that demonstrate significant promise to improve real-time detection of subclinical cancer in the operative setting. CONCLUSIONS There has been an explosion of intraoperative imaging techniques that will become more widespread in the next decade.
Collapse
Affiliation(s)
- Eben L Rosenthal
- *Departments of Surgery and †Radiology, The University of Alabama at Birmingham, Birmingham, AL
| | | | | | | |
Collapse
|
31
|
Abstract
Surgical oncology is experiencing rapid transition in veterinary medicine. Mast cell tumors and soft tissue sarcomas are two of the most common neoplasms in small animal patients. Clinicians should be familiar with the need for staging and the procedures involved in treating patients with these tumors. Clinicians should be comfortable with available adjuvant therapies and when to use them in certain patients.
Collapse
Affiliation(s)
- Brad M Matz
- Surgical Oncology, Department of Clinical Sciences, Auburn University, 1220 Wire Road, Auburn, AL 36849-5540, USA.
| |
Collapse
|
32
|
Boston S, Henderson RA. Role of surgery in multimodal cancer therapy for small animals. Vet Clin North Am Small Anim Pract 2014; 44:855-70. [PMID: 25174903 DOI: 10.1016/j.cvsm.2014.05.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Surgery is a critical component in the treatment of most solid tumors in small animals. Surgery is increasingly combined with adjuvant therapies such as chemotherapy and radiation so surgeons who are treating cancer must have a good understanding of surgical oncology principles, cancer biology, and the roles and potential interactions of surgery, radiation, and chemotherapy. The sequencing plan for these modalities should be determined before treatment is initiated. The surgical oncologist must have a working knowledge of chemotherapy agents and radiation and the effect of these treatments on the ability of tissues to heal and the outcome for the patient.
Collapse
Affiliation(s)
- Sarah Boston
- Department of Small Animal Clinical Sciences, College of Veterinary Medicine, University of Florida, 2015 Southwest 16th Avenue, PO Box 100116, Gainesville, FL 32610, USA.
| | - Ralph A Henderson
- Veterinary Surgical Consulting, 1021 Moores Mill Road, Auburn, AL 36830, USA
| |
Collapse
|
33
|
Imaging primary mouse sarcomas after radiation therapy using cathepsin-activatable fluorescent imaging agents. Int J Radiat Oncol Biol Phys 2013; 86:136-42. [PMID: 23391816 DOI: 10.1016/j.ijrobp.2012.12.007] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2012] [Revised: 12/03/2012] [Accepted: 12/09/2012] [Indexed: 01/23/2023]
Abstract
PURPOSE Cathepsin-activated fluorescent probes can detect tumors in mice and in canine patients. We previously showed that these probes can detect microscopic residual sarcoma in the tumor bed of mice during gross total resection. Many patients with soft tissue sarcoma (STS) and other tumors undergo radiation therapy (RT) before surgery. This study assesses the effect of RT on the ability of cathepsin-activated probes to differentiate between normal and cancerous tissue. METHODS AND MATERIALS A genetically engineered mouse model of STS was used to generate primary hind limb sarcomas that were treated with hypofractionated RT. Mice were injected intravenously with cathepsin-activated fluorescent probes, and various tissues, including the tumor, were imaged using a hand-held imaging device. Resected tumor and normal muscle samples were harvested to assess cathepsin expression by Western blot. Uptake of activated probe was analyzed by flow cytometry and confocal microscopy. Parallel in vitro studies using mouse sarcoma cells were performed. RESULTS RT of primary STS in mice and mouse sarcoma cell lines caused no change in probe activation or cathepsin protease expression. Increasing radiation dose resulted in an upward trend in probe activation. Flow cytometry and immunofluorescence showed that a substantial proportion of probe-labeled cells were CD11b-positive tumor-associated immune cells. CONCLUSIONS In this primary murine model of STS, RT did not affect the ability of cathepsin-activated probes to differentiate between tumor and normal muscle. Cathepsin-activated probes labeled tumor cells and tumor-associated macrophages. Our results suggest that it would be feasible to include patients who have received preoperative RT in clinical studies evaluating cathepsin-activated imaging probes.
Collapse
|