1
|
Bogdanović B, Fagret D, Ghezzi C, Montemagno C. Integrin Targeting and Beyond: Enhancing Cancer Treatment with Dual-Targeting RGD (Arginine-Glycine-Aspartate) Strategies. Pharmaceuticals (Basel) 2024; 17:1556. [PMID: 39598465 PMCID: PMC11597078 DOI: 10.3390/ph17111556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/11/2024] [Accepted: 10/18/2024] [Indexed: 11/29/2024] Open
Abstract
Integrins, an important superfamily of cell adhesion receptors, play an essential role in cancer progression, metastasis, and angiogenesis, establishing them as prime targets for both diagnostic and therapeutic applications. Despite their significant potential, integrin-targeted therapies have faced substantial challenges in clinical trials, including variable efficacy and unmet high expectations. Nevertheless, the consistent expression of integrins on tumor and stromal cells underscores their ongoing relevance and potential. Traditional RGD-based imaging and therapeutic agents have faced limitations, such as inconsistent target expression and rapid systemic clearance, which have reduced their effectiveness. To overcome these challenges, recent research has focused on advancing RGD-based strategies and exploring innovative solutions. This review offers a thorough analysis of the latest developments in the RGD-integrin field, with a particular focus on addressing previous limitations. It delves into new dual-targeting approaches and cutting-edge RGD-based agents designed to improve both tumor diagnosis and therapeutic outcomes. By examining these advancements, this review illuminates new pathways for enhancing the specificity and efficacy of integrin-targeted therapies, paving the way for more effective cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bojana Bogdanović
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Daniel Fagret
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | - Catherine Ghezzi
- INSERM, CHU Grenoble Alpes, Laboratory of Bioclinical Radiopharmaceutics, University Grenoble Alpes, 38000 Grenoble, France; (B.B.); (D.F.); (C.G.)
| | | |
Collapse
|
2
|
Neilio JM, Ginat DT. Emerging Head and Neck Tumor Targeting Contrast Agents for the Purpose of CT, MRI, and Multimodal Diagnostic Imaging: A Molecular Review. Diagnostics (Basel) 2024; 14:1666. [PMID: 39125542 PMCID: PMC11311342 DOI: 10.3390/diagnostics14151666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 07/22/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND The diagnosis and treatment of head and neck tumors present significant challenges due to their infiltrative nature and diagnostic hindrances such as the blood-brain barrier. The intricate anatomy of the head and neck region also complicates the clear identification of tumor boundaries and assessment of tumor characteristics. AIM This review aims to explore the efficacy of molecular imaging techniques that employ targeted contrast agents in head and neck cancer imaging. Head and neck cancer imaging benefits significantly from the combined advantages of CT and MRI. CT excels in providing swift, high-contrast images, enabling the accurate localization of tumors, while MRI offers superior soft tissue resolution, contributing to the detailed evaluation of tumor morphology in this region of the body. Many of these novel contrast agents have integration of dual-modal, triple-modal, or even dual-tissue targeting imaging, which have expanded the horizons of molecular imaging. Emerging contrast agents for the purpose of MRI and CT also include the widely used standards in imaging such as gadolinium and iodine-based agents, respectively, but with peptide, polypeptide, or polymeric functionalizations. Relevance for patients. For patients, the development and use of these targeted contrast agents have potentially significant implications. They benefit from the enhanced accuracy of tumor detection and characterization, which are critical for effective treatment planning. Additionally, these agents offer improved imaging contrast with the added benefit of reduced toxicity and bioaccumulation. The summarization of preclinical nanoparticle research in this review serves as a valuable resource for scientists and students working towards advancing tumor diagnosis and treatment with targeted contrast agents.
Collapse
Affiliation(s)
- Jonathan M. Neilio
- Chicago Medical School, Rosalind Franklin University, North Chicago, IL 60064, USA;
| | - Daniel T. Ginat
- Department of Radiology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
3
|
Sadre Momtaz A, Safarnejad F. 18F-alfatide II internal dosimetry using the ICRP 110 adult reference phantoms and the ICRP 103 tissue weighting factors. Phys Med 2023; 107:102552. [PMID: 36857858 DOI: 10.1016/j.ejmp.2023.102552] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Revised: 02/02/2023] [Accepted: 02/18/2023] [Indexed: 03/03/2023] Open
Abstract
PURPOSE 18F-alfatide II is an arginine-glycine-aspartate (RGD) peptide-based PET tracer with promising imaging properties and pharmacokinetics. This study aims to calculate the absorbed and effective doses of 18F-alfatide II using the ICRP 110 adult reference phantoms and the ICRP 103 tissue weighting factors. METHODS The MIRD method was used in this study to calculate the absorbed dose of organs and tissues. The biokinetic data were taken from a previous study. These data are based on the whole-body PET imaging of mice. RESULTS The results show that the effective dose per unit activity administered of 18F-alfatide II is 1.33E-02 mSv/MBq. The urinary bladder wall receives the highest absorbed dose due to the administration of this radiopharmaceutical. Also, the effective dose of 18F-alfatide II is lower than that of 18F-FDG and some other RGD peptide-based tracers. CONCLUSIONS Dose calculation using ICRP 110 voxelized adult reference phantoms and ICRP 103 tissue weighting factors leads to more realistic and accurate results for 18F-alfatide II compared to the stylized phantoms. The calculated effective dose of 18F-alfatide II in the present study is lower than that of previously published data.
Collapse
Affiliation(s)
- Alireza Sadre Momtaz
- Department of Physics, Faculty of Sciences, University of Guilan, Rasht 41335-1914, Iran.
| | - Farzin Safarnejad
- Department of Physics, Faculty of Sciences, University of Guilan, Rasht 41335-1914, Iran
| |
Collapse
|
4
|
Improved Boron Neutron Capture Therapy Using Integrin αvβ3-Targeted Long-Retention-Type Boron Carrier in a F98 Rat Glioma Model. BIOLOGY 2023; 12:biology12030377. [PMID: 36979069 PMCID: PMC10045558 DOI: 10.3390/biology12030377] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 02/24/2023] [Accepted: 02/25/2023] [Indexed: 03/03/2023]
Abstract
Integrin αvβ3 is more highly expressed in high-grade glioma cells than in normal tissues. In this study, a novel boron-10 carrier containing maleimide-functionalized closo-dodecaborate (MID), serum albumin as a drug delivery system, and cyclic arginine-glycine-aspartate (cRGD) that can target integrin αvβ3 was developed. The efficacy of boron neutron capture therapy (BNCT) targeting integrin αvβ3 in glioma cells in the brain of rats using a cRGD-functionalized MID-albumin conjugate (cRGD-MID-AC) was evaluated. F98 glioma cells exposed to boronophenylalanine (BPA), cRGD-MID-AC, and cRGD + MID were used for cellular uptake and neutron-irradiation experiments. An F98 glioma-bearing rat brain tumor model was used for biodistribution and neutron-irradiation experiments after BPA or cRGD-MID-AC administration. BNCT using cRGD-MID-AC had a sufficient cell-killing effect in vitro, similar to that with BNCT using BPA. In biodistribution experiments, cRGD-MID-AC accumulated in the brain tumor, with the highest boron concentration observed 8 h after administration. Significant differences were observed between the untreated group and BNCT using cRGD-MID-AC groups in the in vivo neutron-irradiation experiments through the log-rank test. Long-term survivors were observed only in BNCT using cRGD-MID-AC groups 8 h after intravenous administration. These findings suggest that BNCT with cRGD-MID-AC is highly selective against gliomas through a mechanism that is different from that of BNCT with BPA.
Collapse
|
5
|
Dobre EG, Surcel M, Constantin C, Ilie MA, Caruntu A, Caruntu C, Neagu M. Skin Cancer Pathobiology at a Glance: A Focus on Imaging Techniques and Their Potential for Improved Diagnosis and Surveillance in Clinical Cohorts. Int J Mol Sci 2023; 24:1079. [PMID: 36674595 PMCID: PMC9866322 DOI: 10.3390/ijms24021079] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2022] [Revised: 01/02/2023] [Accepted: 01/03/2023] [Indexed: 01/08/2023] Open
Abstract
Early diagnosis is essential for completely eradicating skin cancer and maximizing patients' clinical benefits. Emerging optical imaging modalities such as reflectance confocal microscopy (RCM), optical coherence tomography (OCT), magnetic resonance imaging (MRI), near-infrared (NIR) bioimaging, positron emission tomography (PET), and their combinations provide non-invasive imaging data that may help in the early detection of cutaneous tumors and surgical planning. Hence, they seem appropriate for observing dynamic processes such as blood flow, immune cell activation, and tumor energy metabolism, which may be relevant for disease evolution. This review discusses the latest technological and methodological advances in imaging techniques that may be applied for skin cancer detection and monitoring. In the first instance, we will describe the principle and prospective clinical applications of the most commonly used imaging techniques, highlighting the challenges and opportunities of their implementation in the clinical setting. We will also highlight how imaging techniques may complement the molecular and histological approaches in sharpening the non-invasive skin characterization, laying the ground for more personalized approaches in skin cancer patients.
Collapse
Affiliation(s)
- Elena-Georgiana Dobre
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
| | - Mihaela Surcel
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| | - Carolina Constantin
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| | | | - Ana Caruntu
- Department of Oral and Maxillofacial Surgery, “Carol Davila” Central Military Emergency Hospital, 010825 Bucharest, Romania
- Department of Oral and Maxillofacial Surgery, Faculty of Dental Medicine, “Titu Maiorescu” University, 031593 Bucharest, Romania
| | - Constantin Caruntu
- Department of Physiology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Dermatology, “Prof. N.C. Paulescu” National Institute of Diabetes, Nutrition and Metabolic Diseases, 011233 Bucharest, Romania
| | - Monica Neagu
- Faculty of Biology, University of Bucharest, Splaiul Independentei 91-95, 050095 Bucharest, Romania
- Immunology Department, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
- Department of Pathology, Colentina University Hospital, 020125 Bucharest, Romania
| |
Collapse
|
6
|
Shah D, Gehani A, Mahajan A, Chakrabarty N. Advanced Techniques in Head and Neck Cancer Imaging: Guide to Precision Cancer Management. Crit Rev Oncog 2023; 28:45-62. [PMID: 37830215 DOI: 10.1615/critrevoncog.2023047799] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Precision treatment requires precision imaging. With the advent of various advanced techniques in head and neck cancer treatment, imaging has become an integral part of the multidisciplinary approach to head and neck cancer care from diagnosis to staging and also plays a vital role in response evaluation in various tumors. Conventional anatomic imaging (CT scan, MRI, ultrasound) remains basic and focuses on defining the anatomical extent of the disease and its spread. Accurate assessment of the biological behavior of tumors, including tumor cellularity, growth, and response evaluation, is evolving with recent advances in molecular, functional, and hybrid/multiplex imaging. Integration of these various advanced diagnostic imaging and nonimaging methods aids understanding of cancer pathophysiology and provides a more comprehensive evaluation in this era of precision treatment. Here we discuss the current status of various advanced imaging techniques and their applications in head and neck cancer imaging.
Collapse
Affiliation(s)
- Diva Shah
- Senior Consultant Radiologist, Department of Radiodiagnosis, HCG Cancer Centre, Ahmedabad, 380060, Gujarat, India
| | - Anisha Gehani
- Department of Radiology and Imaging Sciences, Tata Medical Centre, New Town, WB 700160, India
| | - Abhishek Mahajan
- Department of Radiology, The Clatterbridge Cancer Centre NHS Foundation Trust, Liverpool, L7 8YA, United Kingdom
| | - Nivedita Chakrabarty
- Department of Radiodiagnosis, Tata Memorial Hospital, Tata Memorial Centre, Homi Bhabha National Institute (HBNI), 400012, Mumbai, India
| |
Collapse
|
7
|
Cao M, Shi E, Wang H, Mao L, Wu Q, Li X, Liang Y, Yang X, Wang Y, Li C. Personalized Targeted Therapeutic Strategies against Oral Squamous Cell Carcinoma. An Evidence-Based Review of Literature. Int J Nanomedicine 2022; 17:4293-4306. [PMID: 36134201 PMCID: PMC9484769 DOI: 10.2147/ijn.s377816] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 09/06/2022] [Indexed: 11/23/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of malignant tumor in the head and neck, with a poor prognosis mainly due to recurrence and metastasis. Classical treatment modalities for OSCC like surgery and radiotherapy have difficulties in dealing with metastatic tumors, and together with chemotherapy, they have major problems related to non-specific cell death. Molecular targeted therapies offer solutions to these problems through not only potentially maximizing the anticancer efficacy but also minimizing the treatment-related toxicity. Among them, the receptor-mediated targeted delivery of anticancer therapeutics remains the most promising one. As OSCC exhibits a heterogeneous nature, selecting the appropriate receptors for targeting is the prerequisite. Hence, we reviewed the OSCC-associated receptors previously used in targeted therapy, focused on their biochemical characteristics and expression patterns, and discussed the application potential in personalized targeted therapy of OSCC. We hope that a better comprehension of this subject will help to provide the fundamental information for OSCC personalized therapeutic planning.
Collapse
Affiliation(s)
- Mingxin Cao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Enyu Shi
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Hanping Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Lujia Mao
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Qiqi Wu
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xinming Li
- Tianjin Stomatological Hospital, School of Medicine, Nankai University, Tianjin Key Laboratory of Oral and Maxillofacial Function Reconstruction, Tianjin, 300041, People's Republic of China
| | - Yanjie Liang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Xiaoying Yang
- Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Yinsong Wang
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China.,Tianjin Key Laboratory of Technologies Enabling Development of Clinical Therapeutics and Diagnostics, School of Pharmacy, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| | - Changyi Li
- School and Hospital of Stomatology, Tianjin Medical University, Tianjin, 300070, People's Republic of China
| |
Collapse
|
8
|
The application value of 18F-Alfatide-RGD PET/CT in the preliminary diagnosis of patients with non-small cell lung cancer. J Radioanal Nucl Chem 2022. [DOI: 10.1007/s10967-022-08496-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2022]
|
9
|
Li D, Li X, Zhao J, Tan F. Advances in nuclear medicine-based molecular imaging in head and neck squamous cell carcinoma. J Transl Med 2022; 20:358. [PMID: 35962347 PMCID: PMC9373390 DOI: 10.1186/s12967-022-03559-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Accepted: 07/25/2022] [Indexed: 12/24/2022] Open
Abstract
Head and neck squamous cell carcinomas (HNSCCs) are often aggressive, making advanced disease very difficult to treat using contemporary modalities, such as surgery, radiation therapy, and chemotherapy. However, targeted therapy, e.g., cetuximab, an epidermal growth factor receptor inhibitor, has demonstrated survival benefit in HNSCC patients with locoregional failure or distant metastasis. Molecular imaging aims at various biomarkers used in targeted therapy, and nuclear medicine-based molecular imaging is a real-time and non-invasive modality with the potential to identify tumor in an earlier and more treatable stage, before anatomic-based imaging reveals diseases. The objective of this comprehensive review is to summarize recent advances in nuclear medicine-based molecular imaging for HNSCC focusing on several commonly radiolabeled biomarkers. The preclinical and clinical applications of these candidate imaging strategies are divided into three categories: those targeting tumor cells, tumor microenvironment, and tumor angiogenesis. This review endeavors to expand the knowledge of molecular biology of HNSCC and help realizing diagnostic potential of molecular imaging in clinical nuclear medicine.
Collapse
Affiliation(s)
- Danni Li
- Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Xuran Li
- Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China
| | - Jun Zhao
- Department of Nuclear Medicine, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Fei Tan
- Shanghai Fourth People's Hospital, and School of Medicine, Tongji University, Shanghai, China. .,The Royal College of Surgeons in Ireland, Dublin, Ireland. .,The Royal College of Surgeons of England, London, UK.
| |
Collapse
|
10
|
Galldiks N, Langen KJ, Albert NL, Law I, Kim MM, Villanueva-Meyer JE, Soffietti R, Wen PY, Weller M, Tonn JC. Investigational PET tracers in neuro-oncology-What's on the horizon? A report of the PET/RANO group. Neuro Oncol 2022; 24:1815-1826. [PMID: 35674736 DOI: 10.1093/neuonc/noac131] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Many studies in patients with brain tumors evaluating innovative PET tracers have been published in recent years, and the initial results are promising. Here, the Response Assessment in Neuro-Oncology (RANO) PET working group provides an overview of the literature on novel investigational PET tracers for brain tumor patients. Furthermore, newer indications of more established PET tracers for the evaluation of glucose metabolism, amino acid transport, hypoxia, cell proliferation, and others are also discussed. Based on the preliminary findings, these novel investigational PET tracers should be further evaluated considering their promising potential. In particular, novel PET probes for imaging of translocator protein and somatostatin receptor overexpression as well as for immune system reactions appear to be of additional clinical value for tumor delineation and therapy monitoring. Progress in developing these radiotracers may contribute to improving brain tumor diagnostics and advancing clinical translational research.
Collapse
Affiliation(s)
- Norbert Galldiks
- Department of Neurology, Faculty of Medicine and University Hospital Cologne, University of Cologne, Kerpener St. 62, 50937 Cologne, Germany.,Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Germany
| | - Karl-Josef Langen
- Institute of Neuroscience and Medicine (INM-3, -4), Research Center Juelich, Juelich, Germany.,Center of Integrated Oncology (CIO), Universities of Aachen, Bonn, Cologne, and Düsseldorf, Germany.,Department of Nuclear Medicine, University Hospital RWTH Aachen, Aachen, Germany
| | - Nathalie L Albert
- Department of Nuclear Medicine, Ludwig Maximilians-University of Munich, Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ian Law
- Department of Clinical Physiology, Nuclear Medicine and PET, Rigshospitalet, University of Copenhagen, Copenhagen, Denmark
| | - Michelle M Kim
- Department of Radiation Oncology, University of Michigan, Ann Arbor, Michigan, USA
| | - Javier E Villanueva-Meyer
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California, USA
| | - Riccardo Soffietti
- Department of Neuro-Oncology, University and City of Health and Science Hospital, Turin, Italy
| | - Patrick Y Wen
- Center for Neuro-Oncology, Dana-Farber/Brigham and Women's Cancer Center, Boston, Massachusetts, USA
| | - Michael Weller
- Department of Neurology, Clinical Neuroscience Center University Hospital and University of Zurich, Zurich, Switzerland
| | - Joerg C Tonn
- Department of Neurosurgery, University Hospital of Munich (LMU), Munich, Germany.,German Cancer Consortium (DKTK), Partner Site Munich, German Cancer Research Center (DKFZ), Heidelberg, Germany
| |
Collapse
|
11
|
Li L, Chen X, Yu J, Yuan S. Preliminary Clinical Application of RGD-Containing Peptides as PET Radiotracers for Imaging Tumors. Front Oncol 2022; 12:837952. [PMID: 35311120 PMCID: PMC8924613 DOI: 10.3389/fonc.2022.837952] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 02/07/2022] [Indexed: 12/15/2022] Open
Abstract
Angiogenesis is a common feature of many physiological processes and pathological conditions. RGD-containing peptides can strongly bind to integrin αvβ3 expressed on endothelial cells in neovessels and several tumor cells with high specificity, making them promising molecular agents for imaging angiogenesis. Although studies of RGD-containing peptides combined with radionuclides, namely, 18F, 64Cu, and 68Ga for positron emission tomography (PET) imaging have shown high spatial resolution and accurate quantification of tracer uptake, only a few of these radiotracers have been successfully translated into clinical use. This review summarizes the RGD-based tracers in terms of accumulation in tumors and adjacent tissues, and comparison with traditional 18F-fluorodeoxyglucose (FDG) imaging. The value of RGD-based tracers for diagnosis, differential diagnosis, tumor subvolume delineation, and therapeutic response prediction is mainly discussed. Very low RGD accumulation, in contrast to high FDG metabolism, was found in normal brain tissue, indicating that RGD-based imaging provides an excellent tumor-to-background ratio for improved brain tumor imaging. However, the intensity of the RGD-based tracers is much higher than FDG in normal liver tissue, which could lead to underestimation of primary or metastatic lesions in liver. In multiple studies, RGD-based imaging successfully realized the diagnosis and differential diagnosis of solid tumors and also the prediction of chemoradiotherapy response, providing complementary rather than similar information relative to FDG imaging. Of most interest, baseline RGD uptake values can not only be used to predict the tumor efficacy of antiangiogenic therapy, but also to monitor the occurrence of adverse events in normal organs. This unique dual predictive value in antiangiogenic therapy may be better than that of FDG-based imaging.
Collapse
Affiliation(s)
- Li Li
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty of Engineering, National University of Singapore, Singapore, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jinming Yu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
| | - Shuanghu Yuan
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital Affiliated to Shandong First Medical University, Jinan, China
- Department of Radiation Oncology, Shandong Cancer Hospital Affiliated to Shandong University, Jinan, China
- Department of Radiation Oncology, The Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
12
|
Alhalhooly L, Confeld MI, Woo SO, Mamnoon B, Jacobson R, Ghosh S, Kim J, Mallik S, Choi Y. Single-Molecule Force Probing of RGD-Binding Integrins on Pancreatic Cancer Cells. ACS APPLIED MATERIALS & INTERFACES 2022; 14:7671-7679. [PMID: 35113515 PMCID: PMC8890904 DOI: 10.1021/acsami.1c23361] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Integrin-targeting arginine-glycine-aspartic acid (RGD)-based nanocarriers have been widely used for tumor imaging, monitoring of tumor development, and delivery of anticancer drugs. However, the thermodynamics of an RGD-integrin formation and dissociation associated with binding dynamics, affinity, and stability remains unclear. Here, we probed the binding strength of the binary complex to live pancreatic cancer cells using single-molecule binding force spectroscopy methods, in which RGD peptides were functionalized on a force probe tip through poly(ethylene glycol) (PEG)-based bifunctional linker molecules. While the density of integrin αV receptors on the cell surface varies more than twofold from cell line to cell line, the individual RGD-integrin complexes exhibited a cell type-independent, monovalent bond strength. The load-dependent bond strength of multivalent RGD-integrin interactions scaled sublinearly with increasing bond number, consistent with the noncooperative, parallel bond model. Furthermore, the multivalent bonds ruptured sequentially either by one or in multiples, and the force strength was comparable to the synchronous rupture force. Comparison of energy landscapes of the bond number revealed a substantial decrease of kinetic off-rates for multivalent bonds, along with the increased width of the potential well and the increased potential barrier height between bound and unbound states, enhancing the stability of the multivalent bonds between them.
Collapse
Affiliation(s)
- Lina Alhalhooly
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Matthew I. Confeld
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sung Oh Woo
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Babak Mamnoon
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Reed Jacobson
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Shrinwanti Ghosh
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Jiha Kim
- Department of Biological Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Sanku Mallik
- Department of Pharmaceutical Sciences, North Dakota State University, Fargo, North Dakota 58108, United State
| | - Yongki Choi
- Department of Physics, North Dakota State University, Fargo, North Dakota 58108, United State
- Molecular and Cellular Biology Program, North Dakota State University, Fargo, North Dakota 58108, United State
- Materials and Nanotechnology Program, North Dakota State University, Fargo, North Dakota 58108, United State
| |
Collapse
|
13
|
Clinical evaluation of kit based Tc-99m-HYNIC-RGD2 for imaging angiogenesis in breast carcinoma patients. Nucl Med Commun 2021; 41:1250-1256. [PMID: 32941401 DOI: 10.1097/mnm.0000000000001282] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
BACKGROUND Radiolabeled RGD peptide can be used for noninvasive in vivo imaging of αvβ3 integrin receptors leading to early detection of tumor cells and hence improving the clinical outcomes. In the present study single vial kit based HYNIC RGD2 was radiolabeled with Tc-99m and evaluated in patients with breast carcinoma. METHODS Radiolabeling was performed via bifunctional chelator method. Tc-99m 1110-2960 MBq (30-80 mCi) was added to the HYNIC-RGD2 vial. The reaction mixture was heated for 20 minutes at 100°C. After performing the quality checks, whole-body planar imaging was performed in 20 patients at 2-2.5 h post i.v. injection of 555-740 MBq (15-20 mCi) of the radiotracer. RESULTS Radiolabeling yield of ≥98% was observed in all the formulations. Quality control tests indicated the suitability of radiopharmaceutical for intravenous administration. Physiological uptake of Tc-99m HYNIC-RGD2 was observed in the nasopharynx, salivary glands, liver, spleen, and intestine. Good uptake of radiotracer was observed in breast lesions of 18 patients. Two patients were observed to be negative. Increased uptake was also seen in metastatic sites in two patients and in lymph nodes in three patients. Scintigraphy findings were in corroboration with pathological observations. CONCLUSION The single vial cold kit based radiolabeling of Tc-99m HYNIC-RGD2 is facile leading to its easy availability. Tc-99m HYNIC-RGD2 is a promising radiopharmaceutical which can be used for the molecular imaging of angiogenesis in breast carcinoma patients.
Collapse
|
14
|
Moody AS, Dayton PA, Zamboni WC. Imaging methods to evaluate tumor microenvironment factors affecting nanoparticle drug delivery and antitumor response. CANCER DRUG RESISTANCE (ALHAMBRA, CALIF.) 2021; 4:382-413. [PMID: 34796317 PMCID: PMC8597952 DOI: 10.20517/cdr.2020.94] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Revised: 01/07/2021] [Accepted: 01/28/2021] [Indexed: 11/24/2022]
Abstract
Standard small molecule and nanoparticulate chemotherapies are used for cancer treatment; however, their effectiveness remains highly variable. One reason for this variable response is hypothesized to be due to nonspecific drug distribution and heterogeneity of the tumor microenvironment, which affect tumor delivery of the agents. Nanoparticle drugs have many theoretical advantages, but due to variability in tumor microenvironment (TME) factors, the overall drug delivery to tumors and associated antitumor response are low. The nanotechnology field would greatly benefit from a thorough analysis of the TME factors that create these physiological barriers to tumor delivery and treatment in preclinical models and in patients. Thus, there is a need to develop methods that can be used to reveal the content of the TME, determine how these TME factors affect drug delivery, and modulate TME factors to increase the tumor delivery and efficacy of nanoparticles. In this review, we will discuss TME factors involved in drug delivery, and how biomedical imaging tools can be used to evaluate tumor barriers and predict drug delivery to tumors and antitumor response.
Collapse
Affiliation(s)
- Amber S. Moody
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - Paul A. Dayton
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Joint Department of Biomedical Engineering, University of North Carolina and North Carolina State University, Chapel Hill, NC 27599, USA
| | - William C. Zamboni
- UNC Eshelman School of Pharmacy, University of North Carolina, Chapel Hill, NC 27599, USA
- UNC Lineberger Comprehensive Cancer Center, Chapel Hill, NC 27599, USA
- Carolina Institute for Nanomedicine, Chapel Hill, NC 27599, USA
| |
Collapse
|
15
|
Lo WL, Lo SW, Chen SJ, Chen MW, Huang YR, Chen LC, Chang CH, Li MH. Molecular Imaging and Preclinical Studies of Radiolabeled Long-Term RGD Peptides in U-87 MG Tumor-Bearing Mice. Int J Mol Sci 2021; 22:ijms22115459. [PMID: 34064291 PMCID: PMC8196871 DOI: 10.3390/ijms22115459] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 05/10/2021] [Accepted: 05/20/2021] [Indexed: 11/18/2022] Open
Abstract
The Arg–Gly–Asp (RGD) peptide shows a high affinity for αvβ3 integrin, which is overexpressed in new tumor blood vessels and many types of tumor cells. The radiolabeled RGD peptide has been studied for cancer imaging and radionuclide therapy. We have developed a long-term tumor-targeting peptide DOTA-EB-cRGDfK, which combines a DOTA chelator, a truncated Evans blue dye (EB), a modified linker, and cRGDfK peptide. The aim of this study was to evaluate the potential of indium-111(111In) radiolabeled DOTA-EB-cRGDfK in αvβ3 integrin-expressing tumors. The human glioblastoma cell line U-87 MG was used to determine the in vitro binding affinity of the radiolabeled peptide. The in vivo distribution of radiolabeled peptides in U-87 MG xenografts was investigated by biodistribution, nanoSPECT/CT, pharmacokinetic and excretion studies. The in vitro competition assay showed that 111In-DOTA-EB-cRGDfK had a significant binding affinity to U-87 MG cancer cells (IC50 = 71.7 nM). NanoSPECT/CT imaging showed 111In-DOTA-EB-cRGDfK has higher tumor uptake than control peptides (111In-DOTA-cRGDfK and 111In-DOTA-EB), and there is still a clear signal until 72 h after injection. The biodistribution results showed significant tumor accumulation (27.1 ± 2.7% ID/g) and the tumor to non-tumor ratio was 22.85 at 24 h after injection. In addition, the pharmacokinetics results indicated that the 111In-DOTA-EB-cRGDfK peptide has a long-term half-life (T1/2λz = 77.3 h) and that the calculated absorbed dose was safe for humans. We demonstrated that radiolabeled DOTA-EB-cRGDfK may be a promising agent for glioblastoma tumor imaging and has the potential as a theranostic radiopharmaceutical.
Collapse
Affiliation(s)
- Wei-Lin Lo
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Shih-Wei Lo
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Su-Jung Chen
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Ming-Wei Chen
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Yuan-Ruei Huang
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Liang-Cheng Chen
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
| | - Chih-Hsien Chang
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
- Department of Biomedical Imaging and Radiological Sciences, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
- Correspondence: (C.-H.C.); (M.-H.L.)
| | - Ming-Hsin Li
- Division of Isotope Application, Institute of Nuclear Energy Research, Taoyuan 32546, Taiwan; (W.-L.L.); (S.-W.L.); (S.-J.C.); (M.-W.C.); (Y.-R.H.); (L.-C.C.)
- Correspondence: (C.-H.C.); (M.-H.L.)
| |
Collapse
|
16
|
Targeted molecular imaging of head and neck squamous cell carcinoma: a window into precision medicine. Chin Med J (Engl) 2021; 133:1325-1336. [PMID: 32404691 PMCID: PMC7289307 DOI: 10.1097/cm9.0000000000000751] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Tumor biomarkers play important roles in tumor growth, invasion, and metastasis. Imaging of specific biomarkers will help to understand different biological activities, thereby achieving precise medicine for each head and neck squamous cell carcinoma (HNSCC) patient. Here, we describe various molecular targets and molecular imaging modalities for HNSCC imaging. An extensive search was undertaken in the PubMed database with the keywords including “HNSCC,” “molecular imaging,” “biomarker,” and “multimodal imaging.” Imaging targets in HNSCC consist of the epidermal growth factor receptor, cluster of differentiation 44 variant 6 (CD44v6), and mesenchymal-epithelial transition factor and integrins. Targeted molecular imaging modalities in HNSCC include optical imaging, ultrasound, magnetic resonance imaging, positron emission tomography, and single-photon emission computed tomography. Making the most of each single imaging method, targeted multimodal imaging has a great potential in the accurate diagnosis and therapy of HNSCC. By visualizing tumor biomarkers at cellular and molecular levels in vivo, targeted molecular imaging can be used to identify specific genetic and metabolic aberrations, thereby accelerating personalized treatment development for HNSCC patients.
Collapse
|
17
|
Makowski MR, Rischpler C, Ebersberger U, Keithahn A, Kasel M, Hoffmann E, Rassaf T, Kessler H, Wester HJ, Nekolla SG, Schwaiger M, Beer AJ. Multiparametric PET and MRI of myocardial damage after myocardial infarction: correlation of integrin αvβ3 expression and myocardial blood flow. Eur J Nucl Med Mol Imaging 2021; 48:1070-1080. [PMID: 32970218 PMCID: PMC8041712 DOI: 10.1007/s00259-020-05034-z] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Increased angiogenesis after myocardial infarction is considered an important favorable prognostic parameter. The αvβ3 integrin is a key mediator of cell-cell and cell-matrix interactions and an important molecular target for imaging of neovasculature and repair processes after MI. Thus, imaging of αvβ3 expression might provide a novel biomarker for assessment of myocardial angiogenesis as a prognostic marker of left ventricular remodeling after MI. Currently, there is limited data available regarding the association of myocardial blood flow and αvβ3 integrin expression after myocardial infarction in humans. METHODS Twelve patients were examined 31 ± 14 days after MI with PET/CT using [18F]Galacto-RGD and [13N]NH3 and with cardiac MRI including late enhancement on the same day. Normal myocardium (remote) and areas of infarction (lesion) were identified on the [18F]Galacto-RGD PET/CT images by correlation with [13N]NH3 PET and cardiac MRI. Lesion/liver-, lesion/blood-, and lesion/remote ratios were calculated. Blood flow and [18F]Galacto-RGD uptake were quantified and correlated for each myocardial segment (AHA 17-segment model). RESULTS In 5 patients, increased [18F]Galacto-RGD uptake was notable within or adjacent to the infarction areas with a lesion/remote ratio of 46% (26-83%; lesion/blood 1.15 ± 0.06; lesion/liver 0.61 ± 0.18). [18F]Galacto-RGD uptake correlated significantly with infarct size (R = 0.73; p = 0.016). Moreover, it correlated significantly with restricted blood flow for all myocardial segments (R = - 0.39; p < 0.0001) and even stronger in severely hypoperfused areas (R = - 0.75; p < 0.0001). CONCLUSION [18F]Galacto-RGD PET/CT allows the visualization and quantification of myocardial αvβ3 expression as a key player in angiogenesis in a subset of patients after MI. αvβ3 expression was more pronounced in patients with larger infarcts and was generally more intense but not restricted to areas with more impaired blood flow, proving that tracer uptake was largely independent of unspecific perfusion effects. Based on these promising results, larger prospective studies are warranted to evaluate the potential of αvβ3 imaging for assessment of myocardial angiogenesis and prediction of ventricular remodeling.
Collapse
Affiliation(s)
- Marcus R Makowski
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany.
- Clinic for Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | | | - Alexandra Keithahn
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Markus Kasel
- Department of Cardiology, Klinikum Bogenhausen, Munich, Germany
| | - Ellen Hoffmann
- Department of Cardiology, Klinikum Bogenhausen, Munich, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study and Center of Integrated Protein Science, Technical University of Munich, Garching, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Stephan G Nekolla
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Nuclear Medicine, University Ulm, Ulm, Germany
| |
Collapse
|
18
|
Notohamiprodjo S, Varasteh Z, Beer AJ, Niu G, Chen X(S, Weber W, Schwaiger M. Tumor Vasculature. Mol Imaging 2021. [DOI: 10.1016/b978-0-12-816386-3.00090-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
|
19
|
Positron Emission Tomography and Molecular Imaging of Head and Neck Malignancies. CURRENT RADIOLOGY REPORTS 2020. [DOI: 10.1007/s40134-020-00366-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
20
|
Durante S, Dunet V, Gorostidi F, Mitsakis P, Schaefer N, Delage J, Prior JO. Head and neck tumors angiogenesis imaging with 68Ga-NODAGA-RGD in comparison to 18F-FDG PET/CT: a pilot study. EJNMMI Res 2020; 10:47. [PMID: 32382869 PMCID: PMC7205972 DOI: 10.1186/s13550-020-00638-w] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2020] [Accepted: 04/24/2020] [Indexed: 12/16/2022] Open
Abstract
Background Angiogenesis plays an important role in head and neck squamous cell carcinoma (HNSCC) progression. This pilot study was designed to compare the distribution of 68Ga-NODAGA-RGD PET/CT for imaging αvβ3 integrins involved in tumor angiogenesis to 18F-FDG PET/CT in patients with HNSCC. Material and methods Ten patients (aged 58.4 ± 8.3 years [range, 44–73 years], 6 males, 4 females) with a total of 11 HNSCC were prospectively enrolled. Activity mapping and standard uptake values (SUV) from both 68Ga-NODAGA-RGD and 18F-FDG PET/CT scans were recorded for primary tumor and compared with the Wilcoxon signed-rank test. The relation between the SUV of both tracers was assessed using the Spearman correlation. Results All HNSCC tumors were visible with both tracers. Quantitative analysis showed higher 18F-FDG SUVmax in comparison to 68Ga-NODAGA-RGD (14.0 ± 6.1 versus 3.9 ± 1.1 g/mL, p = 0.0017) and SUVmean (8.2 ± 3.1 versus 2.0 ± 0.8 g/mL, p = 0.0017). Both 18F-FDG and 68Ga-NODAGA-RGD uptakes were neither correlated with grade, HPV status nor p16 protein expression (p ≥ 0.17). Conclusion All HNSCC tumors were detected with both tracers with higher uptake with 18F-FDG, however. 68Ga-NODAGA-RGD has a different spatial distribution than 18F-FDG bringing different tumor information. Trial registration NCT, NCT02666547. Registered 12.8.2012.
Collapse
Affiliation(s)
- Steve Durante
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon, 46, Lausanne, Switzerland.,Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Vincent Dunet
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon, 46, Lausanne, Switzerland.
| | - François Gorostidi
- Department of Otolaryngology, Head and Neck Surgery, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Periklis Mitsakis
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon, 46, Lausanne, Switzerland
| | - Niklaus Schaefer
- Department of Diagnostic and Interventional Radiology, Lausanne University Hospital and University of Lausanne, Rue du Bugnon, 46, Lausanne, Switzerland
| | - Judith Delage
- Department Pharmacy, Unit of Radiopharmacy, Lausanne University Hospital, Lausanne, Switzerland
| | - John O Prior
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| |
Collapse
|
21
|
Sharma R, Valls PO, Inglese M, Dubash S, Chen M, Gabra H, Montes A, Challapalli A, Arshad M, Tharakan G, Chambers E, Cole T, Lozano-Kuehne JP, Barwick TD, Aboagye EO. [ 18F]Fluciclatide PET as a biomarker of response to combination therapy of pazopanib and paclitaxel in platinum-resistant/refractory ovarian cancer. Eur J Nucl Med Mol Imaging 2020; 47:1239-1251. [PMID: 31754793 PMCID: PMC7101300 DOI: 10.1007/s00259-019-04532-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Accepted: 09/11/2019] [Indexed: 01/02/2023]
Abstract
BACKGROUND Angiogenesis is a driver of platinum resistance in ovarian cancer. We assessed the effect of combination pazopanib and paclitaxel followed by maintenance pazopanib in patients with platinum-resistant/refractory ovarian cancer. Integrins αvβ3 and αvβ5 are both upregulated in tumor-associated vasculature. [18F]Fluciclatide is a novel PET tracer that has high affinity for integrins αvβ3/5, and was used to assess the anti-angiogenic effect of pazopanib. PATIENTS AND METHODS We conducted an open-label, phase Ib study in patients with platinum-resistant/refractory ovarian cancer. Patients received 1 week of single-agent pazopanib (800 mg daily) followed by combination therapy with weekly paclitaxel (80 mg/m2). Following completion of 18 weeks of combination therapy, patients continued with single-agent pazopanib until disease progression. Dynamic [18F]fluciclatide-PET imaging was conducted at baseline and after 1 week of pazopanib. Response (RECIST 1.1), toxicities, and survival outcomes were recorded. Circulating markers of angiogenesis were assessed with therapy. RESULTS Fourteen patients were included in the intention-to-treat analysis. Complete and partial responses were seen in seven patients (54%). Median progression-free survival (PFS) was 10.63 months, and overall survival (OS) was 18.5 months. Baseline [18F]fluciclatide uptake was predictive of long PFS. Elevated baseline circulating angiopoietin and fibroblast growth factor (FGF) were predictive of greater reduction in SUV60,mean following pazopanib. Kinetic modeling of PET data indicated a reduction in K1 and Ki following pazopanib indicating reduced radioligand delivery and retention. CONCLUSIONS Combination therapy followed by maintenance pazopanib is effective and tolerable in platinum-resistant/refractory ovarian cancer. [18F]Fluciclatide-PET uptake parameters predict clinical outcome with pazopanib therapy indicating an anti-angiogenic response.
Collapse
Affiliation(s)
- Rohini Sharma
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK.
| | - Pablo Oriol Valls
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Marianna Inglese
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
- Department of Computer, Control and Management Engineering Antonio Ruberti, University of Rome "La Sapienza", Rome, Italy
| | - Suraiya Dubash
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Michelle Chen
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Hani Gabra
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
- Clinical Discovery Unit, Early Clinical Development, AstraZeneca, Cambridge, UK
| | - Ana Montes
- Department of Medical Oncology, Guy's and St Thomas' NHS Foundation Trust, London, UK
| | | | - Mubarik Arshad
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - George Tharakan
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Ed Chambers
- Division of Diabetes, Endocrinology and Metabolism, Imperial College London, London, UK
| | - Tom Cole
- Department of Medicine, Division of Experimental Medicine, NIHR Imperial Clinical Research Facility, Imperial College London, London, UK
| | - Jingky P Lozano-Kuehne
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| | - Tara D Barwick
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
- Department of Radiology, Imperial College Healthcare NHS Trust, London, UK
| | - Eric O Aboagye
- Department of Surgery and Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| |
Collapse
|
22
|
Imaging angiogenesis in patients with head and neck squamous cell carcinomas by [ 68Ga]Ga-DOTA-E-[c(RGDfK)] 2 PET/CT. Eur J Nucl Med Mol Imaging 2020; 47:2647-2655. [PMID: 32198613 PMCID: PMC7515959 DOI: 10.1007/s00259-020-04766-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/10/2020] [Indexed: 01/07/2023]
Abstract
Purpose Angiogenesis plays an important role in the growth and metastatic spread of solid tumours and is characterised by the expression of integrins on the cell surface of endothelial cells. Radiolabelled RGD peptides specifically target angiogenesis-related αvβ3 integrins, expressed on the activated endothelial cells of sprouting blood vessels. Here, we validated the feasibility of 68Ga[Ga]-DOTA-E-[c(RGDfK)]2 (68Ga-RGD) PET/CT to visualise angiogenesis in patients with oral squamous cell carcinoma (OSCC). Methods Ten patients with OSCC and scheduled for surgical resection including elective neck dissection received an intravenously administration of 68Ga-RGD (42 ± 8 μg; 214 ± 9 MBq). All patients subsequently underwent dynamic (n = 5) or static PET/CT imaging (n = 5) for 60 min or for 4 min/bed position at 30, 60 and 90 min after injection, respectively. Quantitative tracer uptake in tumour lesions was expressed as standardised uptake values (SUV). Additionally, tumour tissue was immunohistochemically stained for αvβ3 integrin to assess the expression pattern. Results 68Ga-RGD tumour accumulation was observed in all patients. At 60 min post injection, tumour SUVmax ranged between 4.0 and 12.7. Tracer accumulation in tumour tissue plateaued at 10 min after injection. Uptake in background tissue did not change over time, resulting in tumour-to-muscle tissue of 6.4 ± 0.7 at 60 min post injection. Conclusions 68Ga-RGD PET/CT of αvβ3 integrin expression in OSCC patients is feasible with adequate tumour-to-background ratios. It will provide more insight in angiogenesis as a hallmark of the head and neck squamous cell carcinomas’ tumour microenvironment. Trial registration https://eudract.ema.europa.eu no. 2015-000917-31 Electronic supplementary material The online version of this article (10.1007/s00259-020-04766-2) contains supplementary material, which is available to authorized users.
Collapse
|
23
|
Rangger C, Haubner R. Radiolabelled Peptides for Positron Emission Tomography and Endoradiotherapy in Oncology. Pharmaceuticals (Basel) 2020; 13:E22. [PMID: 32019275 PMCID: PMC7169460 DOI: 10.3390/ph13020022] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 02/07/2023] Open
Abstract
This review deals with the development of peptide-based radiopharmaceuticals for the use with positron emission tomography and peptide receptor radiotherapy. It discusses the pros and cons of this class of radiopharmaceuticals as well as the different labelling strategies, and summarises approaches to optimise metabolic stability. Additionally, it presents different target structures and addresses corresponding tracers, which are already used in clinical routine or are being investigated in clinical trials.
Collapse
Affiliation(s)
| | - Roland Haubner
- Department of Nuclear Medicine, Medical University of Innsbruck, Anichstrasse 35, 6020 Innsbruck, Austria;
| |
Collapse
|
24
|
Evaluation of Lung Cancer and Neuroendocrine Neoplasm in a Single Scan by Targeting Both Somatostatin Receptor and Integrin αvβ3. Clin Nucl Med 2019; 44:687-694. [DOI: 10.1097/rlu.0000000000002680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
25
|
Diagnostic and Predictive Value of Using RGD PET/CT in Patients with Cancer: A Systematic Review and Meta-Analysis. BIOMED RESEARCH INTERNATIONAL 2019; 2019:8534761. [PMID: 30733968 PMCID: PMC6348803 DOI: 10.1155/2019/8534761] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/07/2018] [Accepted: 12/04/2018] [Indexed: 01/05/2023]
Abstract
The purpose of this study was to assess the diagnostic value of arginine-glycine-aspartic acid (RGD) PET/CT for tumor detection in patients with suspected malignant lesions and to determine the predictive performance of RGD PET/CT in identifying responders. Methods. The PubMed (Medline), EMBASE, Cochrane Library, and Web of Science databases were systematically searched for potentially relevant publications (last updated on July 28th, 2018) reporting the performance of RGD PET in the field of oncology. Pooled sensitivities, specificities, and diagnostic odds ratios (DORs) were calculated for parameters. The areas under the curve (AUCs) and Q⁎ index scores were determined from the constructed summary receiver operating characteristic (SROC) curve. We explored heterogeneity by metaregression. Results. Nine studies, five including 216 patients that determined diagnostic performance and three including 75 patients that determined the predictive value of parameters, met our inclusion criteria. The pooled sensitivity, pooled specificity, DOR, AUC, and Q⁎ index score of RGD PET/CT for the detection of underlying malignancy were 0.85 (0.79-0.89), 0.93 (0.90-0.96), 48.35 (18.95-123.33), 0.9262 (standard error=0.0216), and 0.8606 for SUVmax and 0.86 (0.80-0.91), 0.92 (0.88-0.94), 40.49 (14.16-115.77), 0.9312 (SE=0.0177), and 0.8665 for SUVmean, respectively. The pooled sensitivity, pooled specificity, DOR, AUC, and Q⁎ index score of RGD PET/CT for identifying responders were 0.80 (0.59-0.93), 0.74 (0.60-0.85), 15.76 (4.33-57.32), 0.8682 (0.0539), and 0.7988, respectively, for SUVmax at baseline. Conclusion. The interesting but preliminary data in this meta-analysis demonstrate that RGD PET/CT may be an ideal diagnostic tool for detecting underlying malignancies in patients suspected of having tumors and may be able to efficiently predict short-term outcomes.
Collapse
|
26
|
Collet C, Vucko T, Ariztia J, Karcher G, Pellegrini-Moïse N, Lamandé-Langle S. Fully automated radiosynthesis of [ 18F]fluoro- C-glyco-c(RGDfC): exploiting all the abilities of the AllInOne synthesizer. REACT CHEM ENG 2019. [DOI: 10.1039/c9re00303g] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Fully automated and modular radiosynthesis of [18F]fluoro-C-glyco-RGD conjugate.
Collapse
Affiliation(s)
- Charlotte Collet
- NancycloTEP
- F-54511 Vandoeuvre-lès-Nancy
- France
- Université de Lorraine
- INSERM
| | | | | | - Gilles Karcher
- NancycloTEP
- F-54511 Vandoeuvre-lès-Nancy
- France
- Département de Médecine Nucléaire
- CHRU-Nancy
| | | | | |
Collapse
|
27
|
Meershoek P, van den Berg NS, Brouwer OR, Teertstra HJ, Lange CAH, Valdés-Olmos RA, van der Hiel B, Balm AJM, Klop WMC, van Leeuwen FWB. Three-Dimensional Tumor Margin Demarcation Using the Hybrid Tracer Indocyanine Green- 99mTc-Nanocolloid: A Proof-of-Concept Study in Tongue Cancer Patients Scheduled for Sentinel Node Biopsy. J Nucl Med 2018; 60:764-769. [PMID: 30504140 DOI: 10.2967/jnumed.118.220202] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2018] [Accepted: 10/26/2018] [Indexed: 12/15/2022] Open
Abstract
For radical resection of squamous cell carcinoma of the oral cavity, a tumor-free margin of at least 5 mm is required. Unfortunately, establishing in-depth margins is a surgical conundrum. Knowing that the hybrid sentinel node (SN) tracer indocyanine green (ICG)-99mTc-nanocolloid generates temporary tattoolike markings at the site of administration, we studied the ability to apply this tracer for tumor margin demarcation combined with SN biopsy. Methods: Nineteen patients with clinical T1-T2 oral tongue tumors received the traditional superficial 3 or 4 deposits of ICG-99mTc-nanocolloid (0.1 mL each), and in 12 patients additional deposits were placed deeply using ultrasound guidance (total of 6; 0.07 mL each). SN mapping was performed using lymphoscintigraphy and SPECT/CT. Before and directly after tumor excision, fluorescence imaging was performed to monitor the tracer deposits in the patient (fluorescent deposits were not used to guide the surgical excision). At pathologic examination, primary tumor samples were studied in detail. Results: The number of tracer depositions did not induce a significant difference in the number of SNs visualized (P = 0.836). Reproducible and deep tracer deposition proved to be challenging. The fluorescent nature of ICG-99mTc-nanocolloid supported in vivo and ex vivo identification of the tracer deposits surrounding the tumor. Pathologic examination indicated that in 66.7% (8/12), all fluorescence was observed within the resection margins. Conclusion: This study indicates that tumor margin demarcation combined with SN identification has potential but that some practical challenges need to be overcome if this technique is to mature as a surgical guidance concept. Future studies need to define whether the technology can improve the radical nature of the resections.
Collapse
Affiliation(s)
- Philippa Meershoek
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands.,Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Nynke S van den Berg
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Oscar R Brouwer
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - H Jelle Teertstra
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - Charlotte A H Lange
- Department of Radiology, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands; and
| | - Renato A Valdés-Olmos
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands
| | - Bernies van der Hiel
- Department of Nuclear Medicine, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Alfons J M Balm
- Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - W Martin C Klop
- Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | - Fijs W B van Leeuwen
- Interventional Molecular Imaging Laboratory, Department of Radiology, Leiden University Medical Center, Leiden, The Netherlands .,Department of Head-and-Neck Surgery, Netherlands Cancer Institute-Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| |
Collapse
|
28
|
Debordeaux F, Chansel-Debordeaux L, Pinaquy JB, Fernandez P, Schulz J. What about αvβ3 integrins in molecular imaging in oncology? Nucl Med Biol 2018; 62-63:31-46. [DOI: 10.1016/j.nucmedbio.2018.04.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 03/19/2018] [Accepted: 04/30/2018] [Indexed: 10/17/2022]
|
29
|
Wu J, Wang S, Zhang X, Teng Z, Wang J, Yung BC, Niu G, Zhu H, Lu G, Chen X. 18F-Alfatide II PET/CT for Identification of Breast Cancer: A Preliminary Clinical Study. J Nucl Med 2018; 59:1809-1816. [PMID: 29700127 DOI: 10.2967/jnumed.118.208637] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2018] [Accepted: 04/16/2018] [Indexed: 12/17/2022] Open
Abstract
18F-alfatide II has been proven to have excellent clinical translational potential. In this study, we investigated 18F-alfatide II for identifying breast cancer and compared the performances between 18F-alfatide II and 18F-FDG. Methods: Forty-four female patients with suspected primary breast cancer were recruited. PET/CT images using 18F-alfatide II and 18F-FDG were acquired within 7 d. Tracer uptake in breast lesions was evaluated by visual analysis, and semiquantitative analysis with SUVmax and SUVmean Results: Forty-two breast cancer lesions and 11 benign breast lesions were confirmed by histopathology in 44 patients. Both 18F-alfatide II and 18F-FDG had higher uptake in breast cancer lesions than in benign breast lesions (P < 0.05 for 18F-alfatide II, P < 0.05 for 18F-FDG). The area under the curve of 18F-alfatide II was slightly less than that of 18F-FDG. Both 18F-alfatide II and 18F-FDG had high sensitivity (88.1% vs. 90.5%), high positive predictive value (88.1% vs. 88.4%), moderate specificity (54.5% vs. 54.5%), and moderate negative predictive value (54.5% vs. 60.0%) for differentiating breast cancer from benign breast lesions. By combining 18F-alfatide II and 18F-FDG, the sensitivity and negative predictive value significantly increased to 97.6% and 85.7%, respectively, with positive predictive value slightly increased to 89.1% and no change to the specificity (54.5%). The uptake of 18F-alfatide II (SUVmax: 3.77 ± 1.78) was significantly lower than that of 18F-FDG (SUVmax: 7.37 ± 4.48) in breast cancer lesions (P < 0.05). 18F-alfatide II uptake in triple-negative subtype was significantly lower than that in luminal A and luminal B subtypes. By contrast, human epidermal growth factor receptor-2 (HER-2)-overexpressing subtype had higher 18F-FDG uptake than the other 3 subtypes. There were 8 breast cancer lesions with higher 18F-alfatide II uptake than 18F-FDG uptake, which all had a common characteristic that HER-2 expression was negative and estrogen receptor expression was strongly positive. Conclusion: 18F-alfatide II is suitable for clinical use in breast cancer patients. 18F-alfatide II is of good performance, but not superior to 18F-FDG in identifying breast cancer. 18F-alfatide II may have superiority to 18F-FDG in detecting breast cancer with strongly positive estrogen receptor expression and negative HER-2 expression.
Collapse
Affiliation(s)
- Jiang Wu
- Department of Nuclear Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Shaohua Wang
- Department of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Xianzhong Zhang
- Center for Molecular Imaging and Translational Medicine, Xiamen University, Xiamen, China
| | - Zhaogang Teng
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China; and
| | - Jingjie Wang
- Department of General Surgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Bryant C Yung
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| | - Hong Zhu
- Department of Nuclear Medicine, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China
| | - Guangming Lu
- Department of Medical Imaging, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, China; and
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, Maryland
| |
Collapse
|
30
|
Lobeek D, Franssen GM, Ma MT, Wester HJ, Decristoforo C, Oyen WJG, Boerman OC, Terry SYA, Rijpkema M. In Vivo Characterization of 4 68Ga-Labeled Multimeric RGD Peptides to Image α vβ 3 Integrin Expression in 2 Human Tumor Xenograft Mouse Models. J Nucl Med 2018; 59:1296-1301. [PMID: 29626124 DOI: 10.2967/jnumed.117.206979] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2017] [Accepted: 02/12/2018] [Indexed: 12/11/2022] Open
Abstract
αvβ3 integrins play an important role in angiogenesis and cell migration in cancer and are highly expressed on the activated endothelial cells of newly formed blood vessels. Here, we compare the targeting characteristics of 4 68Ga-labeled multimeric cyclic arginine-glycine-aspartate (RGD)-based tracers in an αvβ3 integrin-expressing tumor model and a tumor model in which αvβ3 integrin is expressed solely on the neovasculature. Methods: Female BALB/c nude mice were subcutaneously injected with SK-RC-52 (αvβ3 integrin-positive) or FaDu (αvβ3 integrin-negative) tumor cells. 68Ga-labeled DOTA-(RGD)2, TRAP-(RGD)3, FSC-(RGD)3, or THP-(RGD)3 was intravenously administered to the mice (0.5 nmol per mouse, 10-20 MBq), followed by small-animal PET/CT imaging and ex vivo biodistribution studies 1 h after injection. Nonspecific uptake of the tracers in both models was determined by coinjecting an excess of unlabeled DOTA-(RGD)2 (50 nmol) along with the radiolabeled tracers. Results: Imaging and biodistribution data showed specific uptake in the tumors for each tracer in both models. Tumor uptake of 68Ga-FSC-(RGD)3 was significantly higher than that of 68Ga-DOTA-(RGD)2, 68Ga-TRAP-(RGD)3, or 68Ga-THP-(RGD)3 in the SK-RC-52 model but not in the FaDu model, in which 68Ga-FSC-(RGD)3 showed significantly higher tumor uptake than 68Ga-TRAP-(RGD)3 Most importantly, differences were also observed in normal tissues and in tumor-to-blood ratios. Conclusion: All tracers showed sufficient targeting of αvβ3 integrin expression to allow for tumor detection. Although the highest tumor uptake was found for 68Ga-FSC-(RGD)3 and 68Ga-THP-(RGD)3 in the SK-RC-52 and FaDu models, respectively, selection of the optimal tracer for specific diagnostic applications also depends on tumor-to-blood ratio and uptake in normal tissues; these factors should therefore also be considered.
Collapse
Affiliation(s)
- Daphne Lobeek
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Gerben M Franssen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Michelle T Ma
- Department of Imaging Chemistry and Biology, King's College London, London, United Kingdom
| | - Hans-Jürgen Wester
- Pharmazeutische Radiochemie, Technische Universität München, Garching, Germany
| | - Clemens Decristoforo
- Department of Nuclear Medicine, Medical University Innsbruck, Innsbruck, Austria; and
| | - Wim J G Oyen
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands.,Institute of Cancer Research and Royal Marsden NHS Trust, Department of Nuclear Medicine, London, United Kingdom
| | - Otto C Boerman
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| | - Samantha Y A Terry
- Department of Imaging Chemistry and Biology, King's College London, London, United Kingdom
| | - Mark Rijpkema
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center Nijmegen, Nijmegen, The Netherlands
| |
Collapse
|
31
|
Cui Y, Liu H, Liang S, Zhang C, Cheng W, Hai W, Yin B, Wang D. The feasibility of 18F-AlF-NOTA-PRGD2 PET/CT for monitoring early response of Endostar antiangiogenic therapy in human nasopharyngeal carcinoma xenograft model compared with 18F-FDG. Oncotarget 2017; 7:27243-54. [PMID: 27029065 PMCID: PMC5053646 DOI: 10.18632/oncotarget.8402] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 03/14/2016] [Indexed: 12/11/2022] Open
Abstract
Purpose Radiolabeled arginine-glycine-aspartic acid (RGD) peptides have been developed for PET imaging of integrin avβ3 in the tumor vasculature, leading to great potential for noninvasively evaluating tumor angiogenesis and monitoring antiangiogenic treatment. The aim of this study was to investigate a novel one-step labeled integrin-targeted tracer, 18F-AlF-NOTA-PRGD2, for PET/CT for detecting tumor angiogenesis and monitoring the early therapeutic efficacy of antiangiogenic agent Endostar in human nasopharyngeal carcinoma (NPC) xenograft model. Experimental design and results Mice bearing NPC underwent 18F-AlF-NOTA-PRGD2 PET/CT at baseline and after 2, 4, 7, and 14 days of consecutive treatment with Endostar or PBS, compared with 18F-FDG PET/CT. Tumors were harvested at all imaging time points for histopathological analysis with H & E and microvessel density (MVD) and integrin avβ3 immunostaining. The maximum percent injected dose per gram of body weight (%ID/gmax) tumor uptake of 18F-AlF-NOTA-PRGD2 PET/CT was significantly lower than that in the control group starting from day 2 (p < 0.01), much earlier and more accurately than that of 18F-FDG PET/CT. Moreover, a moderate linear correlation was observed between tumor MVD and the corresponding tumor uptake of 18F-AlF-NOTA-PRGD2 PET/CT (r = 0.853, p < 0.01). Conclusions 18F-AlF-NOTA-PRGD2 PET/CT can be used for in vivo angiogenesis imaging and monitoring early response to Endostar antiangiogenic treatment in NPC xenograft model, favoring its potential clinical translation.
Collapse
Affiliation(s)
- Yanfen Cui
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Sheng Liang
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Caiyuan Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Weiwei Cheng
- Department of Nuclear Medicine, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Wangxi Hai
- School of Pharmacy, Shanghai Jiao Tong University, Shanghai 200240, China.,Med-X Research Institute, Shanghai Jiao Tong University, Shanghai 200030, China.,Med-X Ruijin Hospital Micro PET/CT Research Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Bing Yin
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China
| |
Collapse
|
32
|
Jackson IM, Scott PJ, Thompson S. Clinical Applications of Radiolabeled Peptides for PET. Semin Nucl Med 2017; 47:493-523. [DOI: 10.1053/j.semnuclmed.2017.05.007] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
33
|
Ahmedah HT, Patterson LH, Shnyder SD, Sheldrake HM. RGD-Binding Integrins in Head and Neck Cancers. Cancers (Basel) 2017; 9:cancers9060056. [PMID: 28587135 PMCID: PMC5483875 DOI: 10.3390/cancers9060056] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2017] [Revised: 05/22/2017] [Accepted: 05/23/2017] [Indexed: 12/13/2022] Open
Abstract
Alterations in integrin expression and function promote tumour growth, invasion, metastasis and neoangiogenesis. Head and neck cancers are highly vascular tumours with a tendency to metastasise. They express a wide range of integrin receptors. Expression of the αv and β1 subunits has been explored relatively extensively and linked to tumour progression and metastasis. Individual receptors αvβ3 and αvβ5 have proved popular targets for diagnostic and therapeutic agents but lesser studied receptors, such as αvβ6, αvβ8, and β1 subfamily members, also show promise. This review presents the current knowledge of integrin expression and function in squamous cell carcinoma of the head and neck (HNSCC), with a particular focus on the arginine-glycine-aspartate (RGD)-binding integrins, in order to highlight the potential of integrins as targets for personalised tumour-specific identification and therapy.
Collapse
Affiliation(s)
- Hanadi Talal Ahmedah
- Radiological Sciences Department, College of Health and Rehabilitation Sciences, Princess Nourah Bint Abdulrahman University, Riyadh 11564, Saudi Arabia.
| | | | - Steven D Shnyder
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| | - Helen M Sheldrake
- Institute of Cancer Therapeutics, University of Bradford, Bradford BD7 1DP, UK.
| |
Collapse
|
34
|
Abstract
The importance of 18F-fluorodesoxyglucose positron-emission tomography (FDG-PET) for the diagnosis of malignant disease is increasing. On one hand, this is due to the high sensitivity of this method, on the other, because the entire body can be examined. FDG-PET can be particularly advantageous for the diagnosis of head and neck tumors, where tumor staging is an important prognostic parameter and essentially determines the therapeutic regimen. This article presents the different possibilities for combined evaluation with PET and computed tomography (CT) for the diagnosis of patients with head and neck cancer. Special focus is placed on primary staging and tumor follow-up, as well as on the role of PET-CT in the diagnosis of patients with cancer of unknown primary origin (CUP). The use of PET-CT for radiotherapy planning and new aspects of PET technology are also discussed.
Collapse
|
35
|
Cui Y, Zhang C, Luo R, Liu H, Zhang Z, Xu T, Zhang Y, Wang D. Noninvasive monitoring of early antiangiogenic therapy response in human nasopharyngeal carcinoma xenograft model using MRI with RGD-conjugated ultrasmall superparamagnetic iron oxide nanoparticles. Int J Nanomedicine 2016; 11:5671-5682. [PMID: 27895477 PMCID: PMC5117895 DOI: 10.2147/ijn.s115357] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Purpose Arginine-glycine-aspartic acid (RGD)-based nanoprobes allow specific imaging of integrin αvβ3, a protein overexpressed during angiogenesis. Therefore, this study applied a novel RGD-coupled, polyacrylic acid (PAA)-coated ultrasmall superparamagnetic iron oxide (USPIO) (referred to as RGD-PAA-USPIO) in order to detect tumor angiogenesis and assess the early response to antiangiogenic treatment in human nasopharyngeal carcinoma (NPC) xenograft model by magnetic resonance imaging (MRI). Materials and methods The binding specificity of RGD-PAA-USPIO with human umbilical vein endothelial cells (HUVECs) was confirmed by Prussian blue staining and transmission electron microscopy in vitro. The tumor targeting of RGD-PAA-USPIO was evaluated in the NPC xenograft model. Later, mice bearing NPC underwent MRI at baseline and after 4 and 14 days of consecutive treatment with Endostar or phosphate-buffered saline (n=10 per group). Results The specific uptake of the RGD-PAA-USPIO nanoparticles was mainly dependent on the interaction between RGD and integrin αvβ3 of HUVECs. The tumor targeting of RGD-PAA-USPIO was observed in the NPC xenograft model. Moreover, the T2 relaxation time of mice in the Endostar-treated group decreased significantly compared with those in the control group both on days 4 and 14, consistent with the immunofluorescence results of CD31 and CD61 (P<0.05). Conclusion This study demonstrated that the magnetic resonance molecular nanoprobes, RGD-PAA-USPIOs, allow noninvasive in vivo imaging of tumor angiogenesis and assessment of the early response to antiangiogenic treatment in NPC xenograft model, favoring its potential clinical translation.
Collapse
Affiliation(s)
- Yanfen Cui
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Caiyuan Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Ran Luo
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Huanhuan Liu
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Zhongyang Zhang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| | - Tianyong Xu
- MR Advanced Application and Research Center, GE Healthcare China, Shanghai, People's Republic of China
| | - Yong Zhang
- MR Advanced Application and Research Center, GE Healthcare China, Shanghai, People's Republic of China
| | - Dengbin Wang
- Department of Radiology, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine
| |
Collapse
|
36
|
Compounds for radionuclide imaging and therapy of malignant foci characterized by the increased angiogenesis. Russ Chem Bull 2016. [DOI: 10.1007/s11172-016-1309-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
37
|
Mena E, Thippsandra S, Yanamadala A, Redy S, Pattanayak P, Subramaniam RM. Molecular Imaging and Precision Medicine in Head and Neck Cancer. PET Clin 2016; 12:7-25. [PMID: 27863568 DOI: 10.1016/j.cpet.2016.08.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The concept of using tumor genomic profiling information has revolutionized personalized cancer treatment. Head and neck (HN) cancer management is being influenced by recent discoveries of activating mutations in epidermal growth factor receptor and related targeted therapies with tyrosine kinase inhibitors, targeted therapies for Kristen Rat Sarcoma, and MET proto-oncogenes. Molecular imaging using PET plays an important role in assessing the biologic behavior of HN cancer with the goal of delivering individualized cancer treatment. This review summarizes recent genomic discoveries in HN cancer and their implications for functional PET imaging in assessing response to targeted therapies, and drug resistance mechanisms.
Collapse
Affiliation(s)
- Esther Mena
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, USA
| | - Shwetha Thippsandra
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, USA
| | - Anusha Yanamadala
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, USA
| | - Siddaling Redy
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, USA
| | - Puskar Pattanayak
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, USA
| | - Rathan M Subramaniam
- Russell H. Morgan Department of Radiology and Radiological Sciences, Johns Hopkins School of Medicine, Johns Hopkins University, 601 North Caroline Street, Baltimore, MD 21287, USA; Department of Radiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-8896, USA; Department of Clinical Sciences, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390-9096, USA; Department of Biomedical Engineering, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8896, USA; Advanced Imaging Research Center, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX, 75390-8896, USA.
| |
Collapse
|
38
|
SPECT and PET imaging of angiogenesis and arteriogenesis in pre-clinical models of myocardial ischemia and peripheral vascular disease. Eur J Nucl Med Mol Imaging 2016; 43:2433-2447. [PMID: 27517840 PMCID: PMC5095166 DOI: 10.1007/s00259-016-3480-8] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 07/28/2016] [Indexed: 01/03/2023]
Abstract
Purpose The extent of neovascularization determines the clinical outcome of coronary artery disease and other occlusive cardiovascular disorders. Monitoring of neovascularization is therefore highly important. This review article will elaborately discuss preclinical studies aimed at validating new nuclear angiogenesis and arteriogenesis tracers. Additionally, we will briefly address possible obstacles that should be considered when designing an arteriogenesis radiotracer. Methods A structured medline search was the base of this review, which gives an overview on different radiopharmaceuticals that have been evaluated in preclinical models. Results Neovascularization is a collective term used to indicate different processes such as angiogenesis and arteriogenesis. However, while it is assumed that sensitive detection through nuclear imaging will facilitate translation of successful therapeutic interventions in preclinical models to the bedside, we still lack specific tracers for neovascularization imaging. Most nuclear imaging research to date has focused on angiogenesis, leaving nuclear arteriogenesis imaging largely overlooked. Conclusion Although angiogenesis is the process which is best understood, there is no scarcity in theoretical targets for arteriogenesis imaging.
Collapse
|
39
|
Shi J, Wang F, Liu S. Radiolabeled cyclic RGD peptides as radiotracers for tumor imaging. BIOPHYSICS REPORTS 2016; 2:1-20. [PMID: 27819026 PMCID: PMC5071373 DOI: 10.1007/s41048-016-0021-8] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/01/2016] [Indexed: 12/19/2022] Open
Abstract
The integrin family comprises 24 transmembrane receptors, each a heterodimeric combination of one of 18α and one of 8β subunits. Their main function is to integrate the cell adhesion and interaction with the extracellular microenvironment with the intracellular signaling and cytoskeletal rearrangement through transmitting signals across the cell membrane upon ligand binding. Integrin αvβ3 is a receptor for the extracellular matrix proteins containing arginine–glycine–aspartic (RGD) tripeptide sequence. The αvβ3 is generally expressed in low levels on the epithelial cells and mature endothelial cells, but it is highly expressed in many solid tumors. The αvβ3 levels correlate well with the potential for tumor metastasis and aggressiveness, which make it an important biological target for development of antiangiogenic drugs, and molecular imaging probes for early tumor diagnosis. Over the last decade, many radiolabeled cyclic RGD peptides have been evaluated as radiotracers for imaging tumors by SPECT or PET. Even though they are called “αvβ3-targeted” radiotracers, the radiolabeled cyclic RGD peptides are also able to bind αvβ5, α5β1, α6β4, α4β1, and αvβ6 integrins, which may help enhance their tumor uptake due to the “increased receptor population.” This article will use the multimeric cyclic RGD peptides as examples to illustrate basic principles for development of integrin-targeted radiotracers and focus on different approaches to maximize their tumor uptake and T/B ratios. It will also discuss important assays for pre-clinical evaluations of the integrin-targeted radiotracers, and their potential applications as molecular imaging tools for noninvasive monitoring of tumor metastasis and early detection of the tumor response to antiangiogenic therapy.
Collapse
Affiliation(s)
- Jiyun Shi
- Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China ; Medical Isotopes Research Center, Peking University, Beijing, 100191 China
| | - Fan Wang
- Interdisciplinary Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101 China ; Medical Isotopes Research Center, Peking University, Beijing, 100191 China
| | - Shuang Liu
- School of Health Sciences, Purdue University, West Lafayette, IN 47907 USA
| |
Collapse
|
40
|
Chen SH, Wang HM, Lin CY, Chang JTC, Hsieh CH, Liao CT, Kang CJ, Yang LY, Yen TC. RGD-K5 PET/CT in patients with advanced head and neck cancer treated with concurrent chemoradiotherapy: Results from a pilot study. Eur J Nucl Med Mol Imaging 2016; 43:1621-9. [PMID: 26922351 DOI: 10.1007/s00259-016-3345-1] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2015] [Accepted: 02/16/2016] [Indexed: 12/15/2022]
Abstract
BACKGROUND/AIM We prospectively investigated the potential usefulness of PET using a new tracer targeting integrin αvβ3 (termed RGD-K5) in patients with head and neck cancer (HNC) undergoing definitive concurrent chemoradiotherapy (CCRT). PATIENTS AND METHODS Newly diagnosed patients with locally advanced HNC scheduled for definitive CCRT were eligible. RDG-K5 PET and FDG PET scans were performed at three different time points (baseline, 2 weeks, and 3 months post-treatment). RESULTS Nine patients completed all of the three scans, whereas two patients withdrew after two scans only. Uptake of both RGD-K5 and FDG generally decreased following CCRT. However, the observed decrease did not differ significantly between complete responders and non-responders. At 3 months post-treatment, the uptake of both RGD-K5 and FDG at the main tumors was significantly lower in those who achieved complete responses than in those with residual tumors. CONCLUSION RGD-K5 PET has the potential to identify patients with incomplete responses to CCRT.
Collapse
Affiliation(s)
- Shih-Hsin Chen
- Department of Nuclear Medicine, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Hung-Ming Wang
- Division of Hematology/Oncology, Department of Internal Medicine, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Chien-Yu Lin
- Department of Radiation Oncology, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Joseph Tung-Chieh Chang
- Department of Radiation Oncology, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Chia-Hsun Hsieh
- Division of Hematology/Oncology, Department of Internal Medicine, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Chun-Ta Liao
- Departments of Otorhinolaryngology, Head and Neck Surgery, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Chung-Jan Kang
- Departments of Otorhinolaryngology, Head and Neck Surgery, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Lan-Yan Yang
- Clinical Trial Center, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333
| | - Tzu-Chen Yen
- Department of Nuclear Medicine, Chang-Gung Memorial Hospital, Linkou, 5, Fu-Shin St., Kwei-Shan Township, TaoYuan County, Taiwan, 333.
| |
Collapse
|
41
|
Withofs N, Hustinx R. Integrin αvβ3 and RGD-based radiopharmaceuticals. MEDECINE NUCLEAIRE-IMAGERIE FONCTIONNELLE ET METABOLIQUE 2016. [DOI: 10.1016/j.mednuc.2015.12.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
42
|
Maschauer S, Gahr S, Gandesiri M, Tripal P, Schneider-Stock R, Kuwert T, Ocker M, Prante O. In vivo monitoring of the anti-angiogenic therapeutic effect of the pan-deacetylase inhibitor panobinostat by small animal PET in a mouse model of gastrointestinal cancers. Nucl Med Biol 2016; 43:27-34. [PMID: 26702784 DOI: 10.1016/j.nucmedbio.2015.10.003] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2015] [Revised: 09/29/2015] [Accepted: 10/16/2015] [Indexed: 12/13/2022]
Abstract
INTRODUCTION Deacetylase inhibitors have recently been established as a novel therapeutic approach to solid and hematologic cancers and have also been demonstrated to possess anti-angiogenic properties. Although these compounds show a good efficacy in vitro and in vivo, no data on monitoring and predicting treatment response are currently available. We therefore investigated the effect of the pan-deacetylase inhibitor panobinostat (LBH589) on gastrointestinal cancer models and the suitability of 2-[(18)F]FGlc-RGD as a specific agent for imaging integrin αvβ3 expression during tumor angiogenesis using small animal positron emission tomography (PET). METHODS The effect of panobinostat on cell viability in vitro was assessed with a label-free impedance based real-time analysis. Nude mice bearing HT29 and HepG2 tumors were treated with daily i.p. injections of 10mg/kg panobinostat for 4 weeks. During this time, tumor size was determined with a calliper and mice were repeatedly subjected to PET imaging. Tumor tissues were analyzed immunohistochemically with a focus on proliferation and endothelial cell markers (Ki-67, Meca-32) and by Western blot applying specific markers of apoptosis. RESULTS In vitro, panobinostat inhibited the proliferation of HepG2 and HT29 cells. Contrary to the situation in HepG2 tumors in vivo, where panobinostat treatment is known to reduce proliferation and vascularization resulting in a decreased tumor growth, HT29 tumors did not show any effect on these parameters. We demonstrated by Western blotting, that panobinostat induced apoptosis in HT29 tumors in vivo. Longitudinal PET imaging studies in HepG2 tumor-bearing mice using 2-[(18)F]FGlc-RGD demonstrated that the standard uptake value (SUVmax) in HepG2 tumors was significantly decreased by 39% at day 7 after treatment. The comparative PET study using HT29 tumor-bearing animals did not reveal any response of the tumors to panobinostat treatment. CONCLUSIONS Small-animal PET imaging using 2-[(18)F]FGlc-RGD was successfully applied to the non-invasive monitoring of the HepG2-tumor response to panobinostat in nude mice early after begin of treatment. Thus, PET imaging of angiogenesis using 2-[(18)F]FGlc-RGD could be a valuable tool to monitor panobinostat therapy in further preclinical studies. ADVANCES IN KNOWLEDGE AND IMPLICATIONS FOR PATIENT CARE When successfully translated to the clinical surrounding, PET imaging of angiogenesis could therefore facilitate therapy planning and monitoring of therapy success with panobinostat in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Simone Maschauer
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University (FAU), Erlangen, Germany
| | - Susanne Gahr
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany
| | - Muktheshwar Gandesiri
- Experimental Tumorpathology, Department of Pathology, Friedrich Alexander University (FAU), Erlangen, Germany
| | - Philipp Tripal
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University (FAU), Erlangen, Germany
| | - Regine Schneider-Stock
- Experimental Tumorpathology, Department of Pathology, Friedrich Alexander University (FAU), Erlangen, Germany
| | - Torsten Kuwert
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University (FAU), Erlangen, Germany
| | - Matthias Ocker
- Department of Medicine 1, University Hospital Erlangen, Erlangen, Germany.
| | - Olaf Prante
- Molecular Imaging and Radiochemistry, Department of Nuclear Medicine, Friedrich Alexander University (FAU), Erlangen, Germany.
| |
Collapse
|
43
|
Chen H, Niu G, Wu H, Chen X. Clinical Application of Radiolabeled RGD Peptides for PET Imaging of Integrin αvβ3. Am J Cancer Res 2016; 6:78-92. [PMID: 26722375 PMCID: PMC4679356 DOI: 10.7150/thno.13242] [Citation(s) in RCA: 223] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 10/06/2015] [Indexed: 12/16/2022] Open
Abstract
Molecular imaging for non-invasive assessment of angiogenesisis is of great interest for clinicians because of the wide-spread application of anti-angiogenic cancer therapeutics. Besides, many other interventions that involve the change of blood vessel/tumor microenvironment would also benefit from such imaging strategies. Of the imaging techniques that target angiogenesis, radiolabeled Arg-Gly-Asp (RGD) peptides have been a major focus because of their high affinity and selectivity for integrin αvβ3--one of the most extensively examined target of angiogenesis. Since the level of integrin αvβ3 expression has been established as a surrogate marker of angiogenic activity, imaging αvβ3 expression can potentially be used as an early indicator of effectiveness of antiangiogenic therapy at the molecular level. In this review, we summarize RGD-based PET tracers that have already been used in clinical trials and intercompared them in terms of radiosynthesis, dosimetry, pharmacokinetics and clinical applications. A perspective of their future use in the clinic is also provided.
Collapse
|
44
|
Kang F, Wang S, Tian F, Zhao M, Zhang M, Wang Z, Li G, Liu C, Yang W, Li X, Wang J. Comparing the Diagnostic Potential of 68Ga-Alfatide II and 18F-FDG in Differentiating Between Non–Small Cell Lung Cancer and Tuberculosis. J Nucl Med 2015; 57:672-7. [DOI: 10.2967/jnumed.115.167924] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2015] [Accepted: 12/04/2015] [Indexed: 12/19/2022] Open
|
45
|
Collet C, Maskali F, Clément A, Chrétien F, Poussier S, Karcher G, Marie PY, Chapleur Y, Lamandé-Langle S. Development of 6-[18F]fluoro-carbohydrate-based prosthetic groups and their conjugation to peptides via click chemistry. J Labelled Comp Radiopharm 2015; 59:54-62. [DOI: 10.1002/jlcr.3362] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2015] [Revised: 11/20/2015] [Accepted: 11/23/2015] [Indexed: 12/27/2022]
Affiliation(s)
- Charlotte Collet
- Université de Lorraine; Vandoeuvre-les-Nancy France
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
| | - Fatiha Maskali
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
| | - Alexandra Clément
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
| | - Françoise Chrétien
- Université de Lorraine; Vandoeuvre-les-Nancy France
- CNRS; UMR 7565; Vandoeuvre-les-Nancy France
| | - Sylvain Poussier
- Université de Lorraine; Vandoeuvre-les-Nancy France
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
| | - Gilles Karcher
- Université de Lorraine; Vandoeuvre-les-Nancy France
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
- Département de Médecine Nucléaire; CHU-Nancy; Vandoeuvre les Nancy France
| | - Pierre-Yves Marie
- Université de Lorraine; Vandoeuvre-les-Nancy France
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
- Département de Médecine Nucléaire; CHU-Nancy; Vandoeuvre les Nancy France
| | - Yves Chapleur
- Université de Lorraine; Vandoeuvre-les-Nancy France
- Nancyclotep, Plateforme d'imagerie moléculaire; Vandoeuvre-les-Nancy France
- CNRS; UMR 7565; Vandoeuvre-les-Nancy France
| | - Sandrine Lamandé-Langle
- Université de Lorraine; Vandoeuvre-les-Nancy France
- CNRS; UMR 7565; Vandoeuvre-les-Nancy France
| |
Collapse
|
46
|
Atallah I, Milet C, Quatre R, Henry M, Reyt E, Coll JL, Hurbin A, Righini C. Role of near-infrared fluorescence imaging in the resection of metastatic lymph nodes in an optimized orthotopic animal model of HNSCC. Eur Ann Otorhinolaryngol Head Neck Dis 2015; 132:337-42. [DOI: 10.1016/j.anorl.2015.08.022] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
|
47
|
Sharma R, Kallur KG, Ryu JS, Parameswaran RV, Lindman H, Avril N, Gleeson FV, Lee JD, Lee KH, O'Doherty MJ, Groves AM, Miller MP, Somer EJ, Coombes CR, Aboagye EO. Multicenter Reproducibility of 18F-Fluciclatide PET Imaging in Subjects with Solid Tumors. J Nucl Med 2015; 56:1855-61. [PMID: 26383153 DOI: 10.2967/jnumed.115.158253] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2015] [Accepted: 06/18/2015] [Indexed: 11/16/2022] Open
Abstract
UNLABELLED Integrins are upregulated on both tumor cells and associated vasculature, where they play an important role in angiogenesis and metastasis. Fluciclatide is an arginine-glycine-aspartic acid peptide with high affinity for αvβ3/αvβ5 integrin, which can be radiolabeled for PET imaging of angiogenesis. Thus, (18)F-fluciclatide is a potential biomarker of therapeutic response to antiangiogenic inhibitors. The aim of this study was to evaluate the reproducibility of (18)F-fluciclatide in multiple solid-tumor types. METHODS Thirty-nine patients underwent PET/CT scanning at 40, 65, and 90 min after injection of (18)F-fluciclatide (maximum, 370 MBq) on 2 separate days (2-9 d apart). Patients did not receive any therapy between PET/CT scans. (18)F-fluciclatide images were reported and quantitative measures of uptake were extracted using the PERCIST methodology. Intrasubject reproducibility of PET uptake in all measurable lesions was evaluated by calculating relative differences in SUV between PET scans for each lesion during the 2 imaging sessions. RESULTS Thirty-nine measurable lesions were detected in 26 patients. Lesion uptake correlated strongly across imaging sessions (r = 0.92, P < 0.05, at 40 min; r = 0.94, P < 0.05, at 65 min; r = 0.94, P < 0.05, at 90 min) with a mean relative difference and SD of the relative difference of 0.006 ± 0.18 at 40 min, 0.003 ± 0.19 at 65 min, and 0.025 ± 0.20 at 90 min. This reflects 95% limits of repeatability of 35%-39% for the difference between the 2 SUV measurements or a variability of 18%-20% in agreement from that observed in well-calibrated multicenter (18)F-FDG studies. CONCLUSION The test-retest reproducibility of (18)F-fluciclatide across multiple tumor types has been measured and shown to be acceptable. This is an important step in the development of this in vivo biomarker to identify and quantify response to antiangiogenic therapy in cancer patients.
Collapse
Affiliation(s)
- Rohini Sharma
- Department of Experimental Medicine, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | | | - Jin S Ryu
- Department of Nuclear Medicine, Asan Medical Center, Seoul, South Korea
| | | | - Henrik Lindman
- Uppsala University Hospital, Department of Oncology, Uppsala, Sweden
| | - Norbert Avril
- Department of Nuclear Medicine, Barts Health, Queen Mary University of London, London, United Kingdom
| | - Fergus V Gleeson
- Department of Radiology, Churchill Hospital, Oxford Radcliffe Hospitals Trust, Oxford, United Kingdom
| | - Jong D Lee
- Department of Nuclear Medicine, Yonsei University Health System, Seoul, Korea
| | - Kyung-Han Lee
- Department of Nuclear Medicine, Samsung Medical Center, Seoul, Korea
| | - Michael J O'Doherty
- PET Imaging Centre, Division of Imaging Sciences and Biomedical Engineering, King's College London, London, United Kingdom
| | - Ashley M Groves
- Institute of Nuclear Medicine, University College London, London, United Kingdom
| | - Matthew P Miller
- GE Healthcare Life Sciences, Imaging R&D, Amersham, United Kingdom; and
| | - Edward J Somer
- GE Healthcare Life Sciences, Imaging R&D, Amersham, United Kingdom; and
| | - Charles R Coombes
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| | - Eric O Aboagye
- Department of Surgery and Cancer, Faculty of Medicine, Imperial College London, Hammersmith Hospital, London, United Kingdom
| |
Collapse
|
48
|
Reyes E. A novel PET tracer for targeted imaging of atherosclerosis. J Nucl Cardiol 2015; 22:1191-4. [PMID: 25721316 DOI: 10.1007/s12350-015-0088-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Accepted: 01/29/2015] [Indexed: 10/23/2022]
Affiliation(s)
- Eliana Reyes
- Royal Brompton and Harefield Hospitals, Sydney Street, London, SW3 6NP, UK.
| |
Collapse
|
49
|
Yu C, Pan D, Mi B, Xu Y, Lang L, Niu G, Yang M, Wan W, Chen X. (18)F-Alfatide II PET/CT in healthy human volunteers and patients with brain metastases. Eur J Nucl Med Mol Imaging 2015; 42:2021-8. [PMID: 26121930 PMCID: PMC4626365 DOI: 10.1007/s00259-015-3118-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 06/14/2015] [Indexed: 01/11/2023]
Abstract
PURPOSE We report the biodistribution and radiation dosimetry of an integrin αvβ3 specific PET tracer (18)F-AlF-NOTA-E[PEG4-c(RGDfk)]2) (denoted as (18)F-Alfatide II). We also assessed the value of (18)F-Alfatide II in patients with brain metastases. METHODS A series of torso (from the skull to the thigh) static images were acquired in five healthy volunteers (3 M, 2 F) at 5, 10, 15, 30, 45, and 60 min after injection of (18)F-Alfatide II (257 ± 48 MBq). Regions of interest (ROIs) were drawn manually, and the time-activity curves (TACs) were obtained for major organs. Nine patients with brain metastases were examined by static PET imaging with (18)F-FDG (5.55 MBq/kg) and (18)F-Alfatide II. RESULTS Injection of (18)F-Alfatide II was well tolerated in all healthy volunteers, with no serious tracer-related adverse events found. (18)F-Alfatide II showed rapid clearance from the blood pool and kidneys. The total effective dose equivalent (EDE) and effective dose (ED) were 0.0277 ± 0.003 mSv/MBq and 0.0198 ± 0.002 mSv/MBq, respectively. The organs with the highest absorbed dose were the kidneys and the spleen. Nine patients with 20 brain metastatic lesions identified by MRI and/or CT were enrolled in this study. All 20 brain lesions were visualized by (18)F-Alfatide II PET, while only ten lesions were visualized by (18)F-FDG, and 13 by CT. CONCLUSION F-Alfatide II is a safe PET tracer with a favorable dosimetry profile. The observed ED suggests that (18)F-Alfatide II is feasible for human studies. (18)F-Alfatide II has potential value in finding brain metastases of different cancers as a biomarker of angiogenesis.
Collapse
Affiliation(s)
- Chunjing Yu
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University (Wuxi No. 4 People's Hospital), Wuxi, China
| | - Donghui Pan
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Baoming Mi
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University (Wuxi No. 4 People's Hospital), Wuxi, China
| | - Yuping Xu
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China
| | - Lixin Lang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Gang Niu
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA
| | - Min Yang
- Key Laboratory of Nuclear Medicine, Ministry of Health, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi, China.
| | - Weixing Wan
- Department of Nuclear Medicine, Affiliated Hospital of Jiangnan University (Wuxi No. 4 People's Hospital), Wuxi, China.
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
50
|
Liu S. Radiolabeled Cyclic RGD Peptide Bioconjugates as Radiotracers Targeting Multiple Integrins. Bioconjug Chem 2015; 26:1413-38. [PMID: 26193072 DOI: 10.1021/acs.bioconjchem.5b00327] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Angiogenesis is a requirement for tumor growth and metastasis. The angiogenic process depends on vascular endothelial cell migration and invasion, and is regulated by various cell adhesion receptors. Integrins are such a family of receptors that facilitate the cellular adhesion to and migration on extracellular matrix proteins in the intercellular spaces and basement membranes. Among 24 members of the integrin family, αvβ3 is studied most extensively for its role in tumor angiogenesis and metastasis. The αvβ3 is expressed at relatively low levels on epithelial cells and mature endothelial cells, but it is highly expressed on the activated endothelial cells of tumor neovasculature and some tumor cells. This restricted expression makes αvβ3 an excellent target to develop antiangiogenic drugs and diagnostic molecular imaging probes. Since αvβ3 is a receptor for extracellular matrix proteins with one or more RGD tripeptide sequence, many radiolabeled cyclic RGD peptides have been evaluated as "αvβ3-targeted" radiotracers for tumor imaging over the past decade. This article will use the dimeric and tetrameric cyclic RGD peptides developed in our laboratories as examples to illustrate basic principles for development of αvβ3-targeted radiotracers. It will focus on different approaches to maximize the radiotracer tumor uptake and tumor/background ratios. This article will also discuss some important assays for preclinical evaluations of integrin-targeted radiotracers. In general, multimerization of cyclic RGD peptides increases their integrin binding affinity and the tumor uptake and retention times of their radiotracers. Regardless of their multiplicity, the capability of cyclic RGD peptides to bind other integrins (namely, αvβ5, α5β1, α6β4, α4β1, and αvβ6) is expected to enhance the radiotracer tumor uptake due to the increased integrin population. The results from preclinical and clinical studies clearly show that radiolabeled cyclic RGD peptides (such as (99m)Tc-3P-RGD2, (18)F-Alfatide-I, and (18)F-Alfatide-II) are useful as the molecular imaging probes for early cancer detection and noninvasive monitoring of the tumor response to antiangiogenic therapy.
Collapse
Affiliation(s)
- Shuang Liu
- School of Health Sciences, Purdue University, 550 Stadium Mall Drive, West Lafayette, Indiana 47907, United States
| |
Collapse
|