1
|
Gromek P, Senkowska Z, Płuciennik E, Pasieka Z, Zhao LY, Gielecińska A, Kciuk M, Kłosiński K, Kałuzińska-Kołat Ż, Kołat D. Revisiting the standards of cancer detection and therapy alongside their comparison to modern methods. World J Methodol 2024; 14:92982. [PMID: 38983668 PMCID: PMC11229876 DOI: 10.5662/wjm.v14.i2.92982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 04/15/2024] [Accepted: 04/28/2024] [Indexed: 06/13/2024] Open
Abstract
In accordance with the World Health Organization data, cancer remains at the forefront of fatal diseases. An upward trend in cancer incidence and mortality has been observed globally, emphasizing that efforts in developing detection and treatment methods should continue. The diagnostic path typically begins with learning the medical history of a patient; this is followed by basic blood tests and imaging tests to indicate where cancer may be located to schedule a needle biopsy. Prompt initiation of diagnosis is crucial since delayed cancer detection entails higher costs of treatment and hospitalization. Thus, there is a need for novel cancer detection methods such as liquid biopsy, elastography, synthetic biosensors, fluorescence imaging, and reflectance confocal microscopy. Conventional therapeutic methods, although still common in clinical practice, pose many limitations and are unsatisfactory. Nowadays, there is a dynamic advancement of clinical research and the development of more precise and effective methods such as oncolytic virotherapy, exosome-based therapy, nanotechnology, dendritic cells, chimeric antigen receptors, immune checkpoint inhibitors, natural product-based therapy, tumor-treating fields, and photodynamic therapy. The present paper compares available data on conventional and modern methods of cancer detection and therapy to facilitate an understanding of this rapidly advancing field and its future directions. As evidenced, modern methods are not without drawbacks; there is still a need to develop new detection strategies and therapeutic approaches to improve sensitivity, specificity, safety, and efficacy. Nevertheless, an appropriate route has been taken, as confirmed by the approval of some modern methods by the Food and Drug Administration.
Collapse
Affiliation(s)
- Piotr Gromek
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Zuzanna Senkowska
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Elżbieta Płuciennik
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
| | - Zbigniew Pasieka
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Lin-Yong Zhao
- Department of General Surgery & Laboratory of Gastric Cancer, State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Adrianna Gielecińska
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz 90-237, Lodzkie, Poland
- Doctoral School of Exact and Natural Sciences, University of Lodz, Lodz 90-237, Lodzkie, Poland
| | - Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Lodz 90-237, Lodzkie, Poland
| | - Karol Kłosiński
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Żaneta Kałuzińska-Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| | - Damian Kołat
- Department of Functional Genomics, Medical University of Lodz, Lodz 90-752, Lodzkie, Poland
- Department of Biomedicine and Experimental Surgery, Medical University of Lodz, Lodz 90-136, Lodzkie, Poland
| |
Collapse
|
2
|
Abdelhafeez AH, Mothi SS, Pio L, Mori M, Santiago TC, McCarville MB, Kaste SC, Pappo AS, Talbot LJ, Murphy AJ, Davidoff AM. Feasibility of indocyanine green-guided localization of pulmonary nodules in children with solid tumors. Pediatr Blood Cancer 2023; 70:e30437. [PMID: 37194488 PMCID: PMC10685698 DOI: 10.1002/pbc.30437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 04/28/2023] [Accepted: 05/02/2023] [Indexed: 05/18/2023]
Abstract
BACKGROUND Clearing all pulmonary metastases is essential for curing pediatric solid tumors. However, intraoperative localization of such pulmonary nodules can be challenging. Therefore, an intraoperative tool that localizes pulmonary metastases is needed to improve diagnostic and therapeutic resections. Indocyanine green (ICG) real-time fluorescence imaging is used for this purpose in adult solid tumors, but its utility in pediatric solid tumors has not been determined. METHODS A single-center, open-label, nonrandomized, prospective clinical trial (NCT04084067) was conducted to assess the ability of ICG to localize pulmonary metastases of pediatric solid tumors. Patients with pulmonary lesions who required resection, either for therapeutic or diagnostic intent, were included. Patients received a 15-minute intravenous infusion of ICG (1.5 mg/kg), and pulmonary metastasectomy was performed the following day. A near-infrared spectroscopy iridium system was optimized to detect ICG, and all procedures were photo-documented and recorded. RESULTS ICG-guided pulmonary metastasectomies were performed in 12 patients (median age: 10.5 years). A total of 79 nodules were visualized, 13 of which were not detected by preoperative imaging. Histologic examination confirmed the following histologies: hepatoblastoma (n = 3), osteosarcoma (n = 2), and one each of rhabdomyosarcoma, Ewing sarcoma, inflammatory myofibroblastic tumor, atypical cartilaginous tumor, neuroblastoma, adrenocortical carcinoma, and papillary thyroid carcinoma. ICG guidance failed to localize pulmonary metastases in five (42%) patients who had inflammatory myofibroblastic tumor, atypical cartilaginous tumor, neuroblastoma, adrenocortical carcinoma, or papillary thyroid carcinoma. CONCLUSIONS ICG-guided identification of pulmonary nodules is not feasible for all pediatric solid tumors. However, it may localize most metastatic hepatic tumors and high-grade sarcomas in children.
Collapse
Affiliation(s)
- Abdelhafeez H. Abdelhafeez
- Departments of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Suraj Sarvode Mothi
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Luca Pio
- Departments of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Motomi Mori
- Department of Biostatistics, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Teresa C. Santiago
- Department of Pathology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - M. Beth McCarville
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Sue C. Kaste
- Department of Diagnostic Imaging, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Alberto S. Pappo
- Department of Oncology, St. Jude Children’s Research Hospital, Memphis, TN, USA
| | - Lindsay J. Talbot
- Departments of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andrew J. Murphy
- Departments of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Andrew M. Davidoff
- Departments of Surgery, St. Jude Children’s Research Hospital, Memphis, TN, USA
- Department of Surgery, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
3
|
Wong LY, Lui NS. Intraoperative Molecular Imaging of Lung Cancer. Thorac Surg Clin 2023; 33:227-232. [PMID: 37414478 DOI: 10.1016/j.thorsurg.2023.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/08/2023]
Abstract
Intraoperative molecular imaging innovations have been propelled by the development of fluorescent contrast agents that specifically target tumor tissues and advanced camera systems that can detect the specified fluorescence. The most promising agent to date is OTL38, a targeted and near-infrared agent that was recently approved by the Food and Drug Administration for intraoperative imaging for lung cancer.
Collapse
Affiliation(s)
- Lye-Yeng Wong
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Falk Building, Stanford, CA 94305, USA. https://twitter.com/LyeYengWongMD
| | - Natalie S Lui
- Department of Cardiothoracic Surgery, Stanford University School of Medicine, 300 Pasteur Drive, Falk Building, Stanford, CA 94305, USA.
| |
Collapse
|
4
|
Azari F, Kennedy G, Bernstein E, Delikatny J, Lee JYK, Kucharczuk J, Low PS, Singhal S. Evaluation of OTL38-Generated Tumor-to-Background Ratio in Intraoperative Molecular Imaging-Guided Lung Cancer Resections. Mol Imaging Biol 2023; 25:85-96. [PMID: 34101106 PMCID: PMC8651846 DOI: 10.1007/s11307-021-01618-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 05/19/2021] [Accepted: 05/23/2021] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Cancer surgery has multiple challenges including localizing small lesions, ensuring negative margins, and identifying synchronous cancers. One of the tools proposed to address these issues is intraoperative molecular imaging (IMI). An important consideration in IMI is the quantification of the tumor fluorescence during the procedure and using that data to add clinical value. Currently, the most commonly cited measure of quantification is the tumor-to-background ratio (TBR). Our goal was to evaluate the clinical value of TBR measured with OTL38 NIR tracer during a lung cancer resection. METHODS Intraoperative data was retrospectively reviewed from a prospectively collected 5-year database. Between 2015 and 2020, 279 patients were included in the study. For standardization, all patients underwent infusion of the same targeted molecular optical contrast agent (OTL38) for lung cancer resections; then, the mean fluorescence intensity of the tumors and background tissues were calculated. To evaluate the clinical efficacy of the TBR calculation, the results were correlated with patient, biologic, tumor, and technological factors. RESULTS For pulmonary surgery, patient factors such as gender, age, smoking history, and time from infusion of OTL38 to surgery did not have any statistical significance in predicting the TBR during surgery. In addition, TBR measurements did not correlate with location of the tumor in the lung (p = 0.123). There was no statistical correlation of preoperative positron emission tomography measurements (standardized uptake value) with intraoperative TBR. However, there was statistically significant negative correlation of in situ TBR measurement and the distance of the lesion from the surface of the organ (p < 0.001). Adenocarcinoma spectrum lesions overall had statistically significant correlation with in situ fluorescence compared to other NSCLC malignancies (p < 0.01) but TBR measurements could not identify histopathologic subtype on univariate analysis (p = 0.089). There was a tendency for in situ fluorescence for moderately and well-differentiated adenocarcinoma spectrum lesions, but this was not statistically significant. When comparing the in situ TBR of benign to malignant nodules in the lung, there was no statistically significant association (p = 0.145). In subset analysis, adenocarcinoma spectrum lesions tend to fluoresce at brighter with OTL38 compared to other histologic subtypes. CONCLUSION In our various iterations, the results of our retrospective analysis did not show that TBR measurements during OTL38-guided surgery provide clinically useful information about the nature of the nodule or cancer. The true value of IMI is in the ability for the surgeon to use the fluorescence to guide the surgeon to the tumor and margins, but that sophisticated quantification of the amount of fluorescence may not have clinical utility.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - Gregory Kennedy
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - Elizabeth Bernstein
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - James Delikatny
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John Y K Lee
- Department of Neurosurgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - John Kucharczuk
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA
| | - Phil S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania Perelman School of Medicine, 3400 Spruce Street, 6 White Building, Philadelphia, PA, 19104, USA.
| |
Collapse
|
5
|
Cui K, Li R, Zhang Y, Qiu Y, Zhao N, Cui Y, Wu W, Liu T, Xiao Z. Molecular Planarization of Raman Probes to Avoid Background Interference for High-Precision Intraoperative Imaging of Tumor Micrometastases and Lymph Nodes. NANO LETTERS 2022; 22:9424-9433. [PMID: 36378880 DOI: 10.1021/acs.nanolett.2c03416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
The intraoperative imaging applications of a large number of Raman probes are hampered by the overlap of their signals with the background Raman signals generated by biological tissues. Here, we describe a molecular planarization strategy for adjusting the Raman shift of these Raman probes to avoid interference. Using this strategy, we modify the backbone of thiophene polymer-poly(3-hexylthiophene) (P3HT), and obtain the adjacent thiophene units planarized polycyclopenta[2,1-b;3,4-b']dithiophene (PCPDT). Compared with P3HT whose signal is disturbed by the Raman signal of lipids in tissues, PCPDT exhibits a 60 cm-1 blueshift in its characteristic signal. Therefore, the PCPDT probe successfully avoids the signal of lipids, and achieves intraoperative imaging of lymph nodes and tumor micrometastasis as small as 0.30 × 0.36 mm. In summary, our study presents a concise molecular planarization strategy for regulating the signal shift of Raman probes, and brings a tunable thiophene polymer probe for high-precision intraoperative Raman imaging.
Collapse
Affiliation(s)
- Kai Cui
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Ruike Li
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Yongming Zhang
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Yuanyuan Qiu
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, People's Republic of China
| | - Na Zhao
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Yanna Cui
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Wenwei Wu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Tize Liu
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
| | - Zeyu Xiao
- Department of Pharmacology and Chemical Biology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, People's Republic of China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200240, People's Republic of China
| |
Collapse
|
6
|
Andreou C, Weissleder R, Kircher MF. Multiplexed imaging in oncology. Nat Biomed Eng 2022; 6:527-540. [PMID: 35624151 DOI: 10.1038/s41551-022-00891-5] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2019] [Accepted: 09/06/2021] [Indexed: 01/24/2023]
Abstract
In oncology, technologies for clinical molecular imaging are used to diagnose patients, establish the efficacy of treatments and monitor the recurrence of disease. Multiplexed methods increase the number of disease-specific biomarkers that can be detected simultaneously, such as the overexpression of oncogenic proteins, aberrant metabolite uptake and anomalous blood perfusion. The quantitative localization of each biomarker could considerably increase the specificity and the accuracy of technologies for clinical molecular imaging to facilitate granular diagnoses, patient stratification and earlier assessments of the responses to administered therapeutics. In this Review, we discuss established techniques for multiplexed imaging and the most promising emerging multiplexing technologies applied to the imaging of isolated tissues and cells and to non-invasive whole-body imaging. We also highlight advances in radiology that have been made possible by multiplexed imaging.
Collapse
Affiliation(s)
- Chrysafis Andreou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, USA.,Department of Electrical and Computer Engineering, University of Cyprus, Nicosia, Cyprus
| | - Ralph Weissleder
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA. .,Department of Systems Biology, Harvard Medical School, Boston, MA, USA.
| | - Moritz F Kircher
- Molecular Pharmacology Program, Sloan Kettering Institute, New York, NY, USA.,Department of Imaging, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA.,Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
7
|
Zhang P, Meng J, Li Y, Yang C, Hou Y, Tang W, McHugh KJ, Jing L. Nanotechnology-enhanced immunotherapy for metastatic cancer. Innovation (N Y) 2021; 2:100174. [PMID: 34766099 PMCID: PMC8571799 DOI: 10.1016/j.xinn.2021.100174] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Accepted: 10/11/2021] [Indexed: 12/14/2022] Open
Abstract
A vast majority of cancer deaths occur as a result of metastasis. Unfortunately, effective treatments for metastases are currently lacking due to the difficulty of selectively targeting these small, delocalized tumors distributed across a variety of organs. However, nanotechnology holds tremendous promise for improving immunotherapeutic outcomes in patients with metastatic cancer. In contrast to conventional cancer immunotherapies, rationally designed nanomaterials can trigger specific tumoricidal effects, thereby improving immune cell access to major sites of metastasis such as bone, lungs, and lymph nodes, optimizing antigen presentation, and inducing a persistent immune response. This paper reviews the cutting-edge trends in nano-immunoengineering for metastatic cancers with an emphasis on different nano-immunotherapeutic strategies. Specifically, it discusses directly reversing the immunological status of the primary tumor, harnessing the potential of peripheral immune cells, preventing the formation of a pre-metastatic niche, and inhibiting the tumor recurrence through postoperative immunotherapy. Finally, we describe the challenges facing the integration of nanoscale immunomodulators and provide a forward-looking perspective on the innovative nanotechnology-based tools that may ultimately prove effective at eradicating metastatic diseases.
Collapse
Affiliation(s)
- Peisen Zhang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Junli Meng
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yingying Li
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Chen Yang
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| | - Yi Hou
- College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Wen Tang
- South China Advanced Institute for Soft Matter Science and Technology, State Key Laboratory of Luminescent Materials and Devices, South China University of Technology, Guangzhou 510640, China
| | - Kevin J McHugh
- Department of Bioengineering, Rice University, 6100 Main Street, MS-142, Houston, TX 77005, USA
| | - Lihong Jing
- Key Laboratory of Colloid, Interface and Chemical Thermodynamics, Institute of Chemistry, Chinese Academy of Sciences, Bei Yi Jie 2, Zhong Guan Cun, Beijing 100190, China
| |
Collapse
|
8
|
Wu Y, Zhang F. Exploiting molecular probes to perform near‐infrared fluorescence‐guided surgery. VIEW 2020. [DOI: 10.1002/viw.20200068] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Affiliation(s)
- Yifan Wu
- Department of Chemistry State Key Laboratory of Molecular Engineering of Polymers Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem Fudan University Shanghai China
| | - Fan Zhang
- Department of Chemistry State Key Laboratory of Molecular Engineering of Polymers Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials and iChem Fudan University Shanghai China
| |
Collapse
|
9
|
Azari F, Kennedy G, Singhal S. Intraoperative Detection and Assessment of Lung Nodules. Surg Oncol Clin N Am 2020; 29:525-541. [PMID: 32883456 DOI: 10.1016/j.soc.2020.06.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Lung cancer is the most frequent cause of cancer-related death worldwide. Despite advances in systemic therapy, the 5-year survival remains humbling at 4% to 17%. For those diagnosed early, surgical therapy can yield potentially curative results. Surgical resection remains a cornerstone of medical care. Success hinges on sound oncologic resection principles. Various techniques can be used to identify pulmonary nodules. A challenge is intraoperative assessment of the surgical specimen to confirm disease localization and ensure an R0 resection. The primary tool is frozen section. Understanding the options available enhances the arsenal of thoracic surgeons and leads to better patient care.
Collapse
Affiliation(s)
- Feredun Azari
- Department of Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 6 White Building, Philadelphia, PA 19104, USA.
| | - Greg Kennedy
- Department of Surgery, Hospital of the University of Pennsylvania, 3400 Spruce Street, 6 White Building, Philadelphia, PA 19104, USA
| | - Sunil Singhal
- Department of Surgery, Division of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, Hospital of the University of Pennsylvania, 3400 Spruce Street, 6 White Building, Philadelphia, PA 19104, USA
| |
Collapse
|
10
|
Pan J, Deng H, Hu S, Xia C, Chen Y, Wang J, Wang Y. Real-time surveillance of surgical margins via ICG-based near-infrared fluorescence imaging in patients with OSCC. World J Surg Oncol 2020; 18:96. [PMID: 32414418 PMCID: PMC7229610 DOI: 10.1186/s12957-020-01874-z] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2020] [Accepted: 05/07/2020] [Indexed: 01/08/2023] Open
Abstract
Background Local recurrence is the main cause of death among patients with oral squamous cell carcinoma (OSCC). This study assessed near-infrared fluorescence (NIF) imaging and spectroscopy to monitor surgical margins intraoperatively for OSCC. Methods Cytological and animal experiments were first performed to confirm the feasibility of monitoring surgical margins with NIF imaging and spectroscopy. Then, 20 patients with OSCC were included in the clinical trials. At 6–8 h after 0.75 mg/kg indocyanine green (ICG) injection, all patients underwent surgery with NIF imaging. During the surgery, both NIF images and quantified fluorescence intensity were acquired to monitor the surgical margins. Results In cytological and animal experiments, the results showed it was feasible to monitor surgical margins with NIF imaging and spectroscopy. Fluorescence was detected in primary tumors in all patients. The fluorescence intensities of the tumor, peritumoral, and normal tissues were 398.863 ± 151.47, 278.52 ± 84.89, and 274.5 ± 100.93 arbitrary units (AUs), respectively (P < 0.05). The SBR of tumor to peritumoral tissue and normal tissues was computed to be 1.45 ± 0.36 and 1.56 ± 0.41, respectively. After primary tumor excision, the wounds showed abnormal fluorescence in four patients (4/20), and residual cancer cells were confirmed by pathological examination in two patients (2/20). Conclusion These findings confirmed the complementary value of NIF imaging during radical tumor resection of OSCC. Before tumor resection, we could utilize the fluorescence margin produced by ICG NIF imaging to determine the surgical margin. Moreover, after tumor blocks were removed, the status of surgical margin could also be evaluated rapidly by ICG NIF imaging of tumor bed and in vitro specimens.
Collapse
Affiliation(s)
- Jiongru Pan
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Han Deng
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Shiqi Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Chengwan Xia
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yongfeng Chen
- Department of Stomatology, The First Affiliated Hospital of Bengbu Medical College, Bengbu, China
| | - Jianquan Wang
- School of Medical Imaging, Bengbu Medical College, Bengbu, China.
| | - Yuxin Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China.
| |
Collapse
|
11
|
Bhatt A, Glehen O. Extent of Peritoneal Resection for Peritoneal Metastases: Looking Beyond a Complete Cytoreduction. Ann Surg Oncol 2020; 27:1458-1470. [PMID: 31965374 DOI: 10.1245/s10434-020-08208-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Indexed: 12/17/2022]
Abstract
Completeness of cytoreduction is one of the most important prognostic factors impacting outcomes of cytoreductive surgery (CRS). To what extent the surrounding normal peritoneum needs to be removed is not known. We hypothesized that the extent of peritoneal resection should be different for different tumors and performed this study to find evidence to support this rationale. To determine the extent of resection of surrounding tissue for any tumor, the mechanisms of tumor development and spread, tumor morphology, the possibility of finding disease in the surrounding normal tissue, and the pattern of lymph node metastases should be known. Surgical resections also depend on patterns of recurrence and the impact of varying extent of resection on survival. We performed a review of literature pertaining to pathways and patterns of peritoneal cancer spread to determine the scientific basis for the extent of peritonectomy. We also reviewed studies comparing less and more extensive peritoneal resection. There is no consensus on the extent of lymphadenectomy required for most PM. Based on this review, we provide recommendations for the extent of peritoneal resection and the extent of lymph node dissection that should be performed for some common peritoneal tumors and identify areas that require further research. We propose that a systematic method of synoptic reporting of pathological specimens of CRS should be developed to capture information regarding the disease distribution within the peritoneal cavity and morphology of PM from different tumors. This can in future be used to establish standard guidelines for such resections.
Collapse
Affiliation(s)
- Aditi Bhatt
- Department of Surgical Oncology, Zydus Hospital, Ahmedabad, India
| | - Olivier Glehen
- Department of Surgical Oncology, Centre Hospitalier Lyon-Sud, Lyon, France.
| |
Collapse
|
12
|
Comparison of a Short Versus Long Stokes Shift Near-Infrared Dye During Intraoperative Molecular Imaging. Mol Imaging Biol 2019; 22:144-155. [DOI: 10.1007/s11307-019-01434-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
13
|
Matsuura Y, Ichinose J, Nakao M, Okumura S, Mun M. Recent fluorescence imaging technology applications of indocyanine green in general thoracic surgery. Surg Today 2019; 50:1332-1342. [PMID: 31664525 DOI: 10.1007/s00595-019-01906-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2019] [Accepted: 09/20/2019] [Indexed: 12/19/2022]
Abstract
Thoracic surgeons perform a wide variety of cancer operations, which are often associated with high morbidity and mortality. Thus, thoracic surgery involves many special challenges that require innovative solutions. The increased utilization of minimally invasive practices, poor overall cancer survival, and significant morbidity of critical operations remain key obstacles to overcome. Fluorescence imaging technology (FIT), involving the implementation of fluorescent dyes and imaging systems, is currently used as an adjunct for general thoracic surgery in many situations and includes sentinel lymph node mapping, pulmonary intersegmental plane identification, pulmonary nodule identification, pulmonary bullous lesion detection, evaluation of the anastomotic perfusion after tracheal surgery, and thoracic duct imaging for postoperative chylothorax. This technology enhances the surgeon's ability to perform operations, and has specific advantages. We review some of the key studies that demonstrate the applications of FIT in the field of general thoracic surgery, focusing on the use of indocyanine green.
Collapse
Affiliation(s)
- Yosuke Matsuura
- Department of Thoracic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan.
| | - Junji Ichinose
- Department of Thoracic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Masayuki Nakao
- Department of Thoracic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Sakae Okumura
- Department of Thoracic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| | - Mingyon Mun
- Department of Thoracic Surgical Oncology, The Cancer Institute Hospital, Japanese Foundation for Cancer Research, 3-8-31, Ariake, Koto-ku, Tokyo, 135-8550, Japan
| |
Collapse
|
14
|
Wang Y, Xie D, Wang Z, Zhang X, Zhang Q, Wang Y, Newton AD, Singhal S, Cai H, Wang Y, Lu Q, Hu Q, Wang Z. Kinetics of indocyanine green: Optimizing tumor to normal tissue fluorescence in image-guided oral cancer surgery applications. Head Neck 2018; 41:1032-1038. [PMID: 30549410 DOI: 10.1002/hed.25541] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2018] [Revised: 08/08/2018] [Accepted: 10/05/2018] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The study aimed to define indocyanine green (ICG) kinetics to determine the optimal ICG dose and surgical time for near-infrared fluorescence-guided oral cancer surgery. METHODS Spectrometer and grayscale digital imaging were used to quantify the ICG kinetics in 12 patients with oral cancer. The fluorescence intensity and signal-to-background ratio (SBR) of tumor and normal tissue were tested at 1, 6, and 24 hours after ICG injection. RESULTS The greatest contrast in the fluorescence intensity between tumor and normal tissue was observed at 6 hours (P < .01), and of three dose groups (0.5, 0.75, and 1.0 mg/kg), 0.75 mg/kg showed the highest SBR (2.06 ± 0.23) after ICG injection. CONCLUSIONS Fluorescence quantification based on spectrometry and grayscale imaging could be effective in determining the optimal ICG dose and surgical time after ICG injection in this cohort of patients with oral cancer.
Collapse
Affiliation(s)
- Yuxin Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Diya Xie
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Ziyang Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Xudong Zhang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Qian Zhang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Yang Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Andrew D Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Huiming Cai
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Yiqing Wang
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Qian Lu
- Department of Biomedical Engineering, College of Engineering and Applied Sciences, Nanjing University, Nanjing, China
| | - Qingang Hu
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| | - Zhiyong Wang
- Department of Oral and Maxillofacial Surgery, Nanjing Stomatological Hospital, Medical School of Nanjing University, Nanjing, China
| |
Collapse
|
15
|
Sengupta S, Krishnan MA, Dudhe P, Reddy RB, Giri B, Chattopadhyay S, Chelvam V. Novel solid-phase strategy for the synthesis of ligand-targeted fluorescent-labelled chelating peptide conjugates as a theranostic tool for cancer. Beilstein J Org Chem 2018; 14:2665-2679. [PMID: 30410628 PMCID: PMC6204756 DOI: 10.3762/bjoc.14.244] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Accepted: 09/28/2018] [Indexed: 12/17/2022] Open
Abstract
In this article, we have successfully designed and demonstrated a novel continuous process for assembling targeting ligands, peptidic spacers, fluorescent tags and a chelating core for the attachment of cytotoxic molecules, radiotracers, nanomaterials in a standard Fmoc solid-phase peptide synthesis in high yield and purity. The differentially protected Fmoc-Lys-(Tfa)-OH plays a vital role in attaching fluorescent tags while growing the peptide chain in an uninterrupted manner. The methodology is versatile for solid-phase resins that are sensitive to mild and strong acidic conditions when acid-sensitive side chain amino protecting groups such as Trt (chlorotrityl), Mtt (4-methyltrityl), Mmt (4-methoxytrityl) are employed to synthesise the ligand targeted fluorescent tagged bioconjugates. Using this methodology, DUPA rhodamine B conjugate (DUPA = 2-[3-(1,3-dicarboxypropyl)ureido]pentanedioic acid), targeting prostate specific membrane antigen (PSMA) expressed on prostate, breast, bladder and brain cancers and pteroate rhodamine B, targeting folate receptor positive cancers such as ovarian, lung, endometrium as well as inflammatory diseases have been synthesized. In vitro studies using LNCaP (PSMA +ve), PC-3 (PSMA −ve, FR −ve) and CHO-β (FR +ve) cell lines and their respective competition experiments demonstrate the specificity of the newly synthesized bioconstructs for future application in fluorescent guided intra-operative imaging.
Collapse
Affiliation(s)
- Sagnik Sengupta
- Discipline of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| | - Mena Asha Krishnan
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| | - Premansh Dudhe
- Discipline of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| | - Ramesh B Reddy
- Discipline of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| | - Bishnubasu Giri
- Discipline of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| | - Sudeshna Chattopadhyay
- Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India.,Discipline of Physics and Discipline of Metallurgy Engineering & Material Sciences, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| | - Venkatesh Chelvam
- Discipline of Chemistry, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India.,Discipline of Biosciences and Biomedical Engineering, Indian Institute of Technology Indore, Khandwa Road, Simrol, Indore 453 552, India
| |
Collapse
|
16
|
Mahalingam SM, Chu H, Liu X, Leamon CP, Low PS. Carbonic Anhydrase IX-Targeted Near-Infrared Dye for Fluorescence Imaging of Hypoxic Tumors. Bioconjug Chem 2018; 29:3320-3331. [PMID: 30185025 DOI: 10.1021/acs.bioconjchem.8b00509] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Use of tumor-targeted fluorescence dyes to help surgeons identify otherwise undetected tumor nodules, decrease the incidence of cancer-positive margins, and facilitate localization of malignant lymph nodes has demonstrated considerable promise for improving cancer debulking surgery. Unfortunately, the repertoire of available tumor-targeted fluorescent dyes does not permit identification of all cancer types, raising the need to develop additional tumor-specific fluorescent dyes to ensure localization of all malignant lesions during cancer surgeries. By comparing the mRNA levels of the hypoxia-induced plasma membrane protein carbonic anhydrase IX (CA IX) in 13 major human cancers with the same mRNA levels in corresponding normal tissues, we document that CA IX constitutes a nearly universal marker for the design of tumor-targeted fluorescent dyes. Motivated by this expression profile, we synthesize two new CA IX-targeted near-infrared (NIR) fluorescent imaging agents and characterize their physical and biological properties both in vitro and in vivo. We report that conjugation of either acetazolamide or 6-aminosaccharin (i.e., two CA-IX-specific ligands) to the NIR fluorescent dye, S0456, via an extended phenolic spacer creates a brightly fluorescent dye that binds CA IX with high affinity and allows rapid visualization of hypoxic regions of solid tumors at depths >1 cm beneath a tissue surface. Taken together, these data suggest that a CA IX-targeted NIR dye can constitute a useful addition to a cocktail of tumor-targeted NIR dyes designed to image all human cancers.
Collapse
Affiliation(s)
| | - Haiyan Chu
- Endocyte Inc. , 3000 Kent Avenue , West Lafayette , Indiana 47906 , United States
| | | | - Christopher P Leamon
- Endocyte Inc. , 3000 Kent Avenue , West Lafayette , Indiana 47906 , United States
| | | |
Collapse
|
17
|
Newton AD, Predina JD, Nie S, Low PS, Singhal S. Intraoperative fluorescence imaging in thoracic surgery. J Surg Oncol 2018; 118:344-355. [PMID: 30098293 DOI: 10.1002/jso.25149] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Accepted: 06/04/2018] [Indexed: 12/19/2022]
Abstract
Intraoperative fluorescence imaging (IFI) can improve real-time identification of cancer cells during an operation. Phase I clinical trials in thoracic surgery have demonstrated that IFI with second window indocyanine green (TumorGlow® ) can identify subcentimeter pulmonary nodules, anterior mediastinal masses, and mesothelioma, while the use of a folate receptor-targeted near-infrared agent, OTL38, can improve the specificity for diagnosing tumors with folate receptor expression. Here, we review the existing preclinical and clinical data on IFI in thoracic surgery.
Collapse
Affiliation(s)
- Andrew D Newton
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Jarrod D Predina
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| | - Shuming Nie
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, Illinois
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania
| |
Collapse
|
18
|
Low PS, Singhal S, Srinivasarao M. Fluorescence-guided surgery of cancer: applications, tools and perspectives. Curr Opin Chem Biol 2018; 45:64-72. [PMID: 29579618 DOI: 10.1016/j.cbpa.2018.03.002] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2018] [Revised: 03/05/2018] [Accepted: 03/08/2018] [Indexed: 12/20/2022]
Abstract
Thousands of patients die each year from residual cancer that remains following cytoreductive surgery. Use of tumor-targeted fluorescent dyes (TTFDs) to illuminate undetected malignant tissue and thereby facilitate its surgical resection shows promise for reducing morbidity and mortality associated with unresected malignant disease. TTFDs can also improve i) detection of recurrent malignant lesions, ii) differentiation of normal from malignant lymph nodes, iii) accurate staging of cancer patients, iv) detection of tumors during robotic/endoscopic surgery (where tumor palpation is no longer possible), and v) preservation of healthy tissue during resection of cancer tissue. Although TTFDs that passively accumulate in a tumor mass provide some tumor contrast, the most encouraging TTFDs in human clinical trials are either enzyme-activated or ligand-targeted to tumor-specific receptors.
Collapse
Affiliation(s)
- Philip S Low
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States.
| | - Sunil Singhal
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania School of Medicine, Philadelphia, PA, United States
| | - Madduri Srinivasarao
- Department of Chemistry and Institute for Drug Discovery, Purdue University, West Lafayette, IN, United States
| |
Collapse
|
19
|
Okusanya OT, Hess NR, Luketich JD, Sarkaria IS. Infrared intraoperative fluorescence imaging using indocyanine green in thoracic surgery. Eur J Cardiothorac Surg 2018; 53:512-518. [PMID: 29029002 DOI: 10.1093/ejcts/ezx352] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2017] [Accepted: 08/30/2017] [Indexed: 01/21/2023] Open
Abstract
Thoracic surgery faces many unique challenges that require innovative solutions. The increase in utilization of minimally invasive practices, poor overall cancer survival and significant morbidity of key operations remain key obstacles to overcome. Intraoperative fluorescence imaging is a process by which fluorescent dyes and imaging systems are used as adjuncts for surgeons in the operating room. Other surgical subspecialists have shown that intraoperative fluorescence imaging can be applied as a practical adjunct to their practices. Thoracic surgeons over the last 15 years have also used intraoperative fluorescence imaging for sentinel lymph node mapping, lung mapping, oesophageal conduit vascular perfusion and lung nodule identification. This review describes some of the key studies that demonstrate the applications of intraoperative near-infrared fluorescence imaging.
Collapse
Affiliation(s)
- Olugbenga T Okusanya
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Nicholas R Hess
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - James D Luketich
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Inderpal S Sarkaria
- Department of Cardiothoracic Surgery, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| |
Collapse
|
20
|
Rogalla S, Joosten SCM, Alam IS, Gambhir SS, Vermesh O. Intraoperative Molecular Imaging in Lung Cancer: The State of the Art and the Future. Mol Ther 2018; 26:338-341. [PMID: 29398484 PMCID: PMC5835150 DOI: 10.1016/j.ymthe.2018.01.013] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Affiliation(s)
- Stephan Rogalla
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sebastiaan C M Joosten
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Israt S Alam
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Sanjiv S Gambhir
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Ophir Vermesh
- Molecular Imaging Program at Stanford (MIPS), Stanford University, Stanford, CA, USA; Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
21
|
Dimastromatteo J, Charles EJ, Laubach VE. Molecular imaging of pulmonary diseases. Respir Res 2018; 19:17. [PMID: 29368614 PMCID: PMC5784614 DOI: 10.1186/s12931-018-0716-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Accepted: 01/05/2018] [Indexed: 12/11/2022] Open
Abstract
Imaging holds an important role in the diagnosis of lung diseases. Along with clinical tests, noninvasive imaging techniques provide complementary and valuable information that enables a complete differential diagnosis. Various novel molecular imaging tools are currently under investigation aimed toward achieving a better understanding of lung disease physiopathology as well as early detection and accurate diagnosis leading to targeted treatment. Recent research on molecular imaging methods that may permit differentiation of the cellular and molecular components of pulmonary disease and monitoring of immune activation are detailed in this review. The application of molecular imaging to lung disease is currently in its early stage, especially compared to other organs or tissues, but future studies will undoubtedly reveal useful pulmonary imaging probes and imaging modalities.
Collapse
Affiliation(s)
- Julien Dimastromatteo
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA USA
| | - Eric J. Charles
- Department of Surgery, University of Virginia, P.O. Box 801359, Charlottesville, VA 22908 USA
| | - Victor E. Laubach
- Department of Surgery, University of Virginia, P.O. Box 801359, Charlottesville, VA 22908 USA
| |
Collapse
|
22
|
Mahalingam SM, Dudkin V, Goldberg S, Klein D, Yi F, Singhal S, O’Neil KT, Low PS. Evaluation of a Centyrin-Based Near-Infrared Probe for Fluorescence-Guided Surgery of Epidermal Growth Factor Receptor Positive Tumors. Bioconjug Chem 2017; 28:2865-2873. [PMID: 28945346 PMCID: PMC11017363 DOI: 10.1021/acs.bioconjchem.7b00566] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Tumor-targeted near-infrared fluorescent dyes have the potential to improve cancer surgery by enabling surgeons to locate and resect more malignant lesions where good visualization tools are required to ensure complete removal of malignant tissue. Although the tumor-targeted fluorescent dyes used in humans to date have been either small organic molecules or high molecular weight antibodies, low molecular weight protein scaffolds have attracted significant attention because they penetrate solid tumors almost as efficiently as small molecules, but can be infinitely mutated to bind almost any antigen. Here we describe the use of a 10 kDa protein scaffold, a Centyrin, to target a near-infrared fluorescent dye to tumors that overexpress the epidermal growth factor receptor (EGFR) for fluorescence-guided surgery (FGS). We have developed and optimized the dose and time required for imaging small tumor burdens with minimal background fluorescence in real-time fluorescence-guided surgery of EGFR-expressing tumor xenografts in murine models. We demonstrate that the Centyrin-near-infrared dye conjugate (CNDC) binds selectively to human EGFR+ cancer cells with an EC50 of 2 nM, localizes to EGFR+ tumor xenografts in athymic nude mice and that uptake of the dye in xenografts is significantly reduced when EGFR are blocked by preinjection of excess unlabeled Centyrin. Taken together, these data suggest that CNDCs can be used for intraoperative identification and surgical removal of EGFR-expressing lesions and that Centyrins targeted to other tumor-specific antigens should prove similarly useful in fluorescence guided surgery of cancer. In addition, we demonstrate that the CNDC is detected in the NIR region of the spectrum and can be utilized for fluorescence-guided surgery (FGS). In addition, we propose that with its eventual complete clearance from EGFR-negative tissues and its quantitative retention in the tumor mass for >24 h, a Centyrin-targeted NIR dye should provide excellent tumor contrast when injected at least 6-8 h before initiation of cancer surgery in human patients.
Collapse
Affiliation(s)
- Sakkarapalayam M. Mahalingam
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| | - Vadim Dudkin
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Shalom Goldberg
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Donna Klein
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Fang Yi
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania 19104, United States
| | - Karyn T. O’Neil
- Janssen Research & Development, 1400 McKean Road, Springhouse PA 19477, United States
| | - Philip S. Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana 47907, United States
- Institute for Drug Discovery, Purdue University, West Lafayette, Indiana 47907, United States
| |
Collapse
|
23
|
Hekman MCH, Boerman OC, Bos DL, Massuger LFAG, Weil S, Grasso L, Rybinski KA, Oosterwijk E, Mulders PFA, Rijpkema M. Improved Intraoperative Detection of Ovarian Cancer by Folate Receptor Alpha Targeted Dual-Modality Imaging. Mol Pharm 2017; 14:3457-3463. [PMID: 28826214 PMCID: PMC6150714 DOI: 10.1021/acs.molpharmaceut.7b00464] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
![]()
Complete resection of tumor lesions
in advanced stage ovarian cancer patients is of utmost importance,
since the extent of residual disease after surgery strongly affects
survival. Intraoperative imaging may be useful to improve surgery
in these patients. Farletuzumab is a humanized IgG1 antibody that
specifically recognizes the folate receptor alpha (FRα). Labeled
with a radiolabel and a fluorescent dye, farletuzumab may be used
for the intraoperative detection of ovarian cancer lesions. The current
aim is to demonstrate the feasibility of FRα-targeted dual-modality
imaging using 111In-farletuzumab-IRDye800CW in an intraperitoneal
ovarian cancer model. Biodistribution studies were performed 3 days
after injection of 3, 10, 30, or 100 μg of 111In-farletuzumab-IRDye800CW
in mice with subcutaneous IGROV-1 tumors (5 mice per group). In mice
with intraperitoneal IGROV-1 tumors the nonspecific uptake of 111In-farletuzumab-IRDye800CW was determined by coinjecting
an excess of unlabeled farletuzumab. MicroSPECT/CT and fluorescence
imaging were performed 3 days after injection of 10 μg of 111In-farletuzumab-IRDye800CW. FRα expression in tumors
was determined immunohistochemically. Optimal tumor-to-blood-ratios
(3.4–3.7) were obtained at protein doses up to 30 μg.
Multiple intra-abdominal tumor lesions were clearly visualized by
microSPECT/CT, while uptake in normal tissues was limited. Fluorescence
imaging was used to visualize and guide resection of superficial tumors.
Coinjection of an excess of unlabeled farletuzumab significantly decreased
tumor uptake of 111In-farletuzumab-IRDye800CW (69.4 ±
27.6 versus 18.3 ± 2.2% ID/g, p < 0.05).
Immunohistochemical analyses demonstrated that the radioactive and
fluorescent signal corresponded with FRα-expressing tumor lesions.
FRα-targeted SPECT/fluorescence imaging using 111In-farletuzumab-IRDye800CW can be used to detect ovarian cancer in vivo and could be a valuable tool for enhanced intraoperative
tumor visualization in patients with intraperitoneal metastases of
ovarian cancer.
Collapse
Affiliation(s)
- Marlène C H Hekman
- Department of Radiology and Nuclear Medicine, Radboudumc , Nijmegen 6525 GA, Netherlands.,Department of Urology, Radboudumc , Nijmegen 6525 GA, Netherlands
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboudumc , Nijmegen 6525 GA, Netherlands
| | - Desirée L Bos
- Department of Radiology and Nuclear Medicine, Radboudumc , Nijmegen 6525 GA, Netherlands
| | | | - Susan Weil
- Morphotek , Exton, Pennsylvania 19341, United States
| | - Luigi Grasso
- Morphotek , Exton, Pennsylvania 19341, United States
| | | | | | | | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboudumc , Nijmegen 6525 GA, Netherlands
| |
Collapse
|
24
|
A Glimpse of the Future With Intraoperative Molecular Imaging. Ann Surg 2017; 266:e45. [PMID: 28604423 DOI: 10.1097/sla.0000000000002320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
25
|
Kennedy GT, Newton A, Predina J, Singhal S. Intraoperative near-infrared imaging of mesothelioma. Transl Lung Cancer Res 2017; 6:279-284. [PMID: 28713673 DOI: 10.21037/tlcr.2017.05.01] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Though difficult to achieve, complete resection of malignant pleural mesothelioma is paramount to improving patient survival. Surgeons have traditionally been limited to using visual inspection and manual palpation to locate and remove cancerous tissue. However, intraoperative molecular imaging (IMI) is a promising new technology in surgery. Molecular imaging utilizes a fluorescent tracer that selectively accumulates in cancer cells. An imaging device is then used to detect and augment the fluorescent signal emitted from the fluorescent cancer cells. Our group and others have demonstrated that molecular imaging with either indocyanine green (ICG) or a folate receptor alpha (FRα) targeted fluorophore can accurately identify a number of intrathoracic malignancies. Early studies of intraoperative imaging have suggested its efficacy for malignant pleural mesothelioma. In a murine model of mesothelioma, intraoperative imaging was found to have sensitivity of 87% and specificity of 83%. In a pilot human study, eight patients with biopsy-proven epithelial malignant pleural mesothelioma were administered 5 mg/kg of intravenous ICG injection 24 h prior to resection. The following day, a near-infrared (NIR) imaging device was used to detect tumor fluorescence intraoperatively. After what was believed to be complete tumor excision, the wound bed was re-imaged for residual fluorescence indicative of retained tumor. When residual fluorescence was detected, additional tissue was resected, if feasible, and specimens were sent for pathologic correlation. In all cases, intraoperative fluorescence localized to mesothelioma deposits which were confirmed on final pathology. Following resection, fluorescence was confirmed ex vivo with a mean tumor-to-background ratio (TBR) of 3.2 (IQR: 2.9-3.4). It is hoped that this technology will improve outcomes for mesothelioma patients by allowing for a more complete oncologic resection.
Collapse
Affiliation(s)
- Gregory Thomas Kennedy
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Andrew Newton
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Jarrod Predina
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania School of Medicine, Philadelphia, PA, USA
| |
Collapse
|
26
|
Xia L, Venegas OG, Predina JD, Singhal S, Guzzo TJ. Intraoperative Molecular Imaging for Post-Chemotherapy Robot-Assisted Laparoscopic Resection of Seminoma Metastasis: A Case Report. Clin Genitourin Cancer 2017; 15:e61-e64. [PMID: 27793611 PMCID: PMC10998540 DOI: 10.1016/j.clgc.2016.09.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Accepted: 09/16/2016] [Indexed: 11/30/2022]
Affiliation(s)
- Leilei Xia
- Division of Urology, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Ollin G Venegas
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Division of Thoracic Surgery and Thoracic Surgery Research Laboratory, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Jarrod D Predina
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Division of Thoracic Surgery and Thoracic Surgery Research Laboratory, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Sunil Singhal
- Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Division of Thoracic Surgery and Thoracic Surgery Research Laboratory, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA
| | - Thomas J Guzzo
- Division of Urology, Department of Surgery, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA.
| |
Collapse
|
27
|
Keating JJ, Runge JJ, Singhal S, Nims S, Venegas O, Durham AC, Swain G, Nie S, Low PS, Holt DE. Intraoperative near-infrared fluorescence imaging targeting folate receptors identifies lung cancer in a large-animal model. Cancer 2016; 123:1051-1060. [PMID: 28263385 DOI: 10.1002/cncr.30419] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2016] [Revised: 09/14/2016] [Accepted: 10/04/2016] [Indexed: 12/19/2022]
Abstract
BACKGROUND Complete tumor resection is the most important predictor of patient survival with non-small cell lung cancer. Methods for intraoperative margin assessment after lung cancer excision are lacking. This study evaluated near-infrared (NIR) intraoperative imaging with a folate-targeted molecular contrast agent (OTL0038) for the localization of primary lung adenocarcinomas, lymph node sampling, and margin assessment. METHODS Ten dogs with lung cancer underwent either video-assisted thoracoscopic surgery or open thoracotomy and tumor excision after an intravenous injection of OTL0038. Lungs were imaged with an NIR imaging device both in vivo and ex vivo. The wound bed was re-imaged for retained fluorescence suspicious for positive tumor margins. The tumor signal-to-background ratio (SBR) was measured in all cases. Next, 3 human patients were enrolled in a proof-of-principle study. Tumor fluorescence was measured both in situ and ex vivo. RESULTS All canine tumors fluoresced in situ (mean Fluoptics SBR, 5.2 [range, 2.7-8.1]; mean Karl Storz SBR 1.9 [range, 1.4-2.6]). In addition, the fluorescence was consistent with tumor margins on pathology. Three positive lymph nodes were discovered with NIR imaging. Also, a positive retained tumor margin was discovered upon NIR imaging of the wound bed. Human pulmonary adenocarcinomas were also fluorescent both in situ and ex vivo (mean SBR, > 2.0). CONCLUSIONS NIR imaging can identify lung cancer in a large-animal model. In addition, NIR imaging can discriminate lymph nodes harboring cancer cells and also bring attention to a positive tumor margin. In humans, pulmonary adenocarcinomas fluoresce after the injection of the targeted contrast agent. Cancer 2017;123:1051-60. © 2016 American Cancer Society.
Collapse
Affiliation(s)
- Jane J Keating
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Precision Surgery, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jeffrey J Runge
- Center for Precision Surgery, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sunil Singhal
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Precision Surgery, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Sarah Nims
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Precision Surgery, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Ollin Venegas
- Department of Surgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Center for Precision Surgery, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amy C Durham
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Gary Swain
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Shuming Nie
- Department of Biomedical Engineering, Emory University, Atlanta, Georgia.,Department of Chemistry, Emory University, Atlanta, Georgia
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, Indiana
| | - David E Holt
- Center for Precision Surgery, Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania.,Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
28
|
Newton AD, Kennedy GT, Predina JD, Low PS, Singhal S. Intraoperative molecular imaging to identify lung adenocarcinomas. J Thorac Dis 2016; 8:S697-S704. [PMID: 28066672 DOI: 10.21037/jtd.2016.09.50] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intraoperative molecular imaging is a promising new technology with numerous applications in lung cancer surgery. Accurate identification of small nodules and assessment of tumor margins are two challenges in pulmonary resections for cancer, particularly with increasing use of video-assisted thoracoscopic surgery (VATS). One potential solution to these problems is intraoperative use of a fluorescent contrast agent to improve detection of cancer cells. This technology requires both a targeted fluorescent dye that will selectively accumulate in cancer cells and a specialized imaging system to detect the cells. In several studies, we have shown that intraoperative imaging with indocyanine green (ICG) can be used to accurately identify indeterminate pulmonary nodules. The use of a folate-tagged fluorescent molecule targeted to the folate receptor-α (FRα) further improves the sensitivity and specificity of detecting lung adenocarcinomas. We have demonstrated this technology can be used as an "optical biopsy" to differentiate adenocarcinoma versus other histological subtypes of pulmonary nodules. This strategy has potential applications in assessing bronchial stump margins, identifying synchronous or metachronous lesions, and rapidly assessing lymph nodes for lung adenocarcinoma.
Collapse
Affiliation(s)
- Andrew D Newton
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA, USA
| | - Gregory T Kennedy
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA, USA
| | - Jarrod D Predina
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA, USA
| | - Philip S Low
- Department of Chemistry, Purdue University, West Lafayette, IN, USA
| | - Sunil Singhal
- Department of Surgery, University of Pennsylvania Perelman School of Medicine, Pennsylvania, PA, USA
| |
Collapse
|
29
|
Armato SG, Blyth KG, Keating JJ, Katz S, Tsim S, Coolen J, Gudmundsson E, Opitz I, Nowak AK. Imaging in pleural mesothelioma: A review of the 13th International Conference of the International Mesothelioma Interest Group. Lung Cancer 2016; 101:48-58. [PMID: 27794408 DOI: 10.1016/j.lungcan.2016.09.003] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2016] [Accepted: 09/05/2016] [Indexed: 12/17/2022]
Abstract
Imaging plays an important role in the detection, diagnosis, staging, response assessment, and surveillance of malignant pleural mesothelioma. The etiology, biology, and growth pattern of mesothelioma present unique challenges for each modality used to capture various aspects of this disease. Clinical implementation of imaging techniques and information derived from images continue to evolve based on active research in this field worldwide. This paper summarizes the imaging-based research presented orally at the 2016 International Conference of the International Mesothelioma Interest Group (iMig) in Birmingham, United Kingdom, held May 1-4, 2016. Presented topics included intraoperative near-infrared imaging of mesothelioma to aid the assessment of resection completeness, an evaluation of tumor enhancement improvement with increased time delay between contrast injection and image acquisition in standard clinical magnetic resonance imaging (MRI) scans, the potential of early contrast enhancement analysis to provide MRI with a role in mesothelioma detection, the differentiation of short- and long-term survivors based on MRI tumor volume and histogram analysis, the response-assessment potential of hemodynamic parameters derived from dynamic contrast-enhanced computed tomography (DCE-CT) scans, the correlation of CT-based tumor volume with post-surgical tumor specimen weight, and consideration of the need to update the mesothelioma tumor response assessment paradigm.
Collapse
Affiliation(s)
- Samuel G Armato
- Department of Radiology, The University of Chicago, Chicago, Illinois, USA.
| | - Kevin G Blyth
- Department of Respiratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK and Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Jane J Keating
- Department of Surgery, University of Pennsylvania Perelman School of Medicine and Philadelphia Veterans Affairs Medical Center, Philadelphia, PA, USA; Center for Precision Surgery, Abramson Cancer Center, University of Pennsylvania Pearlman School of Medicine, Philadelphia, PA, USA
| | - Sharyn Katz
- Department of Radiology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Selina Tsim
- Department of Respiratory Medicine, Queen Elizabeth University Hospital, Glasgow, UK and Institute of Infection, Immunity & Inflammation, University of Glasgow, Glasgow, UK
| | - Johan Coolen
- Department of Radiology, University Hospitals Leuven, Belgium
| | | | - Isabelle Opitz
- Division of Thoracic Surgery, University Hospital Zurich, Zurich, Switzerland
| | - Anna K Nowak
- School of Medicine and Pharmacology and National Centre for Asbestos Related Diseases, University of Western Australia, Perth, Western Australia and Department of Medical Oncology, Sir Charles Gairdner Hospital, Perth, Western Australia, Australia
| |
Collapse
|
30
|
Zhang H, Desai P, Koike Y, Houghton J, Carlin S, Tandon N, Touijer K, Weber WA. Dual-Modality Imaging of Prostate Cancer with a Fluorescent and Radiogallium-Labeled Gastrin-Releasing Peptide Receptor Antagonist. J Nucl Med 2016; 58:29-35. [PMID: 27516447 DOI: 10.2967/jnumed.116.176099] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Accepted: 07/12/2016] [Indexed: 11/16/2022] Open
Abstract
Gastrin-releasing peptide (GRP) receptors (GRPr) are frequently overexpressed in human prostate cancer, and radiolabeled GRPr affinity ligands have shown promise for in vivo imaging of prostate cancer with PET. The goal of this study was to develop a dual-modality imaging probe that can be used for noninvasive PET imaging and optical imaging of prostate cancer. METHODS We designed and synthesized an IRDye 650 and DOTA-conjugated GRPr antagonist, HZ220 (DOTA-Lys(IRDye 650)-PEG4-[D-Phe6, Sta13]-BN(6-14)NH2), by reacting DOTA-Lys-PEG4-[D-Phe6, Sta13]-BN(6-14)NH2 (HZ219) with IRDye 650 N-hydroxysuccinimide (NHS) ester. Receptor-specific binding of gallium-labeled HZ220 was characterized in PC-3 prostate cancer cells (PC-3), and tumor uptake in mice was imaged with PET/CT and fluorescence imaging. Receptor binding affinity, in vivo tumor uptake, and biodistribution were compared with the GRPr antagonists HZ219, DOTA-PEG4-[D-Phe6, Sta13]-BN(6-14)NH2 (DOTA-AR), and DOTA-(4-amino-1-carboxymethyl-piperidine)-[D-Phe6, Sta13]-BN(6-14)NH2 (DOTA-RM2). RESULTS After hydrophilic-lipophilic balance cartridge purification, 68Ga-HZ220 was obtained with a radiochemical yield of 56% ± 8% (non-decay-corrected), and the radiochemical purity was greater than 95%. Ga-HZ220 had a lower affinity for GRPr (inhibitory concentration of 50% [IC50], 21.4 ± 7.4 nM) than Ga-DOTA-AR (IC50, 0.48 ± 0.18 nM) or Ga-HZ219 (IC50, 0.69 ± 0.18 nM). Nevertheless, 68Ga-HZ220 had an in vivo tumor accumulation similar to 68Ga-DOTA-AR (4.63 ± 0.31 vs. 4.07 ± 0.29 percentage injected activity per mL [%IA/mL] at 1 h after injection) but lower than that of 68Ga-DOTA-RM2 (10.4 ± 0.4 %IA/mL). The tumor uptake of 68Ga-HZ220 was blocked significantly with an excessive amount of GRP antagonists. IVIS spectrum imaging also visualized PC-3 xenografts in vivo and ex vivo with a high-contrast ratio. Autoradiography and fluorescent-based microscopic imaging with 68Ga-HZ220 consistently colocated the expression of GRPr. 68Ga-HZ220 displayed a higher kidney uptake than both 68Ga-DOTA-AR and 68Ga-DOTA-RM2 (16.9 ± 6.5 vs. 4.48 ± 1.63 vs. 5.01 ± 2.29 %IA/mL). CONCLUSION 68Ga-HZ220 is a promising bimodal ligand for noninvasive PET imaging and intraoperative optical imaging of GRPr-expressing malignancies. Bimodal nuclear/fluorescence imaging may not only improve cancer detection and guide surgical resections, but also improve our understanding of the uptake of GRPr ligands on the cellular level.
Collapse
Affiliation(s)
- Hanwen Zhang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Pooja Desai
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Yusuke Koike
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York.,Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Jacob Houghton
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sean Carlin
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Nidhi Tandon
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Karim Touijer
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, New York; and
| | - Wolfgang A Weber
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York .,Molecular Pharmacology and Chemistry Program, Memorial Sloan Kettering Cancer Center, New York, New York
| |
Collapse
|
31
|
Keating J, Tchou J, Okusanya O, Fisher C, Batiste R, Jiang J, Kennedy G, Nie S, Singhal S. Identification of breast cancer margins using intraoperative near-infrared imaging. J Surg Oncol 2016; 113:508-14. [PMID: 26843131 DOI: 10.1002/jso.24167] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2015] [Accepted: 12/27/2015] [Indexed: 12/19/2022]
Abstract
BACKGROUND AND OBJECTIVES Current methods of intraoperative breast cancer margin assessment are labor intensive, not fully reliable, and time consuming; therefore novel strategies are necessary. We hypothesized that near infrared (NIR) intraoperative molecular imaging using systemic indocyanine green (ICG) would be helpful in discerning tumor margins. METHODS A mammary cancer cell line, 4T1, was used to establish tumors in mouse flanks (n = 60). Tumors were excised 24 hr after intravenous ICG. Assessment of residual tumor in the wound bed was performed using a combination of NIR imaging and traditional method (by visual inspection and palpation) versus traditional method alone. Next we performed a clinical trial to evaluate the role of NIR imaging after systemic ICG for the margin assessment of 12 patients undergoing breast-conserving surgery. RESULTS Traditional margin assessment identified 30% of positive margins while NIR imaging identified 90% of positive margins. In our clinical trial, all tumors were detected by NIR imaging and there was fluorescent evidence of residual tumor in the tumor bed in 6 of the 12 patients. None of these patients had positive margins on pathology. CONCLUSIONS Systemic ICG reliably accumulates in breast cancers in murine models as well as human breast cancer. While NIR imaging is helpful for detection of retained tumor margins in our animal model, intraoperative imaging for precise margin detection will need further refinement before clinical value can be obtained. J. Surg. Oncol. 2016;113:508-514. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Jane Keating
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Julia Tchou
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Olugbenga Okusanya
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Carla Fisher
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Rebecca Batiste
- Department of Pathology, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Jack Jiang
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Gregory Kennedy
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| | - Shuming Nie
- Departments of Biomedical Engineering and Chemistry, Emory University, 1364 Clifton Road, Atlanta, Georgia
| | - Sunil Singhal
- Department of Thoracic Surgery, University of Pennsylvania, Perelman School of Medicine, 3400 Spruce Street, Philadelphia, Pennsylvania
| |
Collapse
|
32
|
Quantification of tumor fluorescence during intraoperative optical cancer imaging. Sci Rep 2015; 5:16208. [PMID: 26563091 PMCID: PMC4643322 DOI: 10.1038/srep16208] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 09/29/2015] [Indexed: 12/18/2022] Open
Abstract
Intraoperative optical cancer imaging is an emerging technology in which surgeons employ fluorophores to visualize tumors, identify tumor-positive margins and lymph nodes containing metastases. This study compares instrumentation to measure tumor fluorescence. Three imaging systems (Spectropen, Glomax, Flocam) measured and quantified fluorescent signal-to-background ratios (SBR) in vitro, murine xenografts, tissue phantoms and clinically. Evaluation criteria included the detection of small changes in fluorescence, sensitivity of signal detection at increasing depths and practicality of use. In vitro, spectroscopy was superior in detecting incremental differences in fluorescence than luminescence and digital imaging (Ln[SBR] = 6.8 ± 0.6, 2.4 ± 0.3, 2.6 ± 0.1, p = 0.0001). In fluorescent tumor cells, digital imaging measured higher SBRs than luminescence (6.1 ± 0.2 vs. 4.3 ± 0.4, p = 0.001). Spectroscopy was more sensitive than luminometry and digital imaging in identifying murine tumor fluorescence (SBR = 41.7 ± 11.5, 5.1 ± 1.8, 4.1 ± 0.9, p = 0.0001), and more sensitive than digital imaging at detecting fluorescence at increasing depths (SBR = 7.0 ± 3.4 vs. 2.4 ± 0.5, p = 0.03). Lastly, digital imaging was the most practical and least time-consuming. All methods detected incremental differences in fluorescence. Spectroscopy was the most sensitive for small changes in fluorescence. Digital imaging was the most practical considering its wide field of view, background noise filtering capability, and sensitivity to increasing depth.
Collapse
|