1
|
Wang G, Pan Y, Zheng L, Zhang X, Liu H, Xu Y, Zhang W, Luan X, Liu X, Xu X, Wu S, Ma G, Kan Y, Zhang J, Wang R, Yang J. Monitoring Sorafenib Resistance and Efficacy in Hepatocellular Carcinoma Using [ 18F]Alfatide II and [ 18F]Fluorodeoxyglucose Positron Emission Tomography. Mol Pharm 2025; 22:2088-2097. [PMID: 39988972 DOI: 10.1021/acs.molpharmaceut.4c01218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Integrin αvβ3 expression is associated with sorafenib resistance in hepatocellular carcinoma (HCC). Therefore, monitoring its expression in HCC may serve as a valuable indicator of the efficacy of sorafenib treatment. In this study, longitudinal positron emission tomography (PET) was performed to assess [18F]Alfatide II and [18F]fluorodeoxyglucose ([18F]FDG) as suitable probes for evaluating the treatment efficacy of sorafenib in a Huh-7 human (HCC) xenograft model. Huh-7 tumor cells were used to establish both normal and sorafenib-resistant cell lines, and xenograft models were developed. The mice were categorized into four groups based on the cell type and treatment: normal nontreatment, normal treatment, sorafenib-resistant nontreatment, and sorafenib-resistant treatment. Huh-7 tumor mice received intragastric injections of sorafenib (30 mg/kg/day) or vehicle for 15 consecutive days. Tumor size and weight were assessed throughout the study. Longitudinal microPET/computed tomography (CT) scans with [18F]Alfatide II and [18F]FDG were acquired to quantitatively measure angiogenesis on days -2, 3, 7, and 14 and metabolism on days -1, 4, 8, and 15 following therapy initiation. The tumor uptake (ID%/gmean) of each probe was calculated. No significant difference in [18F]FDG uptake was observed between the normal and sorafenib-resistant groups (P = 0.452); however, [18F]Alfatide II uptake differed significantly between the two groups (P < 0.001). Sorafenib successfully inhibited normal Huh-7 tumor growth, inducing significant differences in tumor size 9 days after sorafenib treatment (P < 0.05). The uptake of [18F]Alfatide II in the tumor lesions changed significantly on day 14 (P = 0.001). However, no change was observed in the uptake of [18F]FDG (P > 0.05). The PET imaging data of [18F]Alfatide II and [18F]FDG were validated through ex vivo immunohistochemistry analysis targeting integrin αvβ3, VEGF, and GULT-1. [18F]Alfatide II PET was more effective in monitoring sorafenib resistance and therapeutic efficacy in the Huh-7 human HCC xenograft model than [18F]FDG.
Collapse
Affiliation(s)
- Guanyun Wang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Yue Pan
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Graduate School, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Lingling Zheng
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Xiaojun Zhang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Huanhuan Liu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
- Graduate School, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Yanfeng Xu
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Wenwen Zhang
- Department of Gastroenterology and Hepatology, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiaohui Luan
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiaojie Liu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Xiaodan Xu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Shina Wu
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Guangyu Ma
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Ying Kan
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| | - Jinming Zhang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Ruimin Wang
- Department of Nuclear Medicine, The First Medical Center, Chinese PLA General Hospital, 28 Fuxing Road, Haidian District, Beijing 100853, China
| | - Jigang Yang
- Nuclear Medicine Department, Beijing Friendship Hospital, Capital Medical University, 95 Yong'an Road, Xicheng District, Beijing 100050, China
| |
Collapse
|
2
|
Butt A, Bach H. Advancements in nanotechnology for diagnostics: a literature review, part II: advanced techniques in nuclear and optical imaging. Nanomedicine (Lond) 2025; 20:183-206. [PMID: 39670826 PMCID: PMC11730800 DOI: 10.1080/17435889.2024.2439778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024] Open
Abstract
Modern molecular imaging routes, such as nuclear imaging and optical imaging, derive significant advantages from nanoparticles, where multimodality use and multipurpose are key benefits. Nanoparticles also showcase benefits over traditional imaging agents in nuclear and optical imaging, including improved resolution, penetration, and specificity. The goal of this literature review was to explore recent advancements in nanomaterials within these molecular imaging techniques to expand on the current state of nanomedicine in these modalities. This review derives findings from relevant reviews, original research papers, in-human clinical trials, and patents in the literature. Au- and Fe oxide-based nanosystems are just as ubiquitous within more modern modalities due to their multimodal diagnostic and therapeutic potential. It is also repeatedly highlighted in the literature, patents, and clinical trials that the use of nanoparticles, specifically in multimodal imaging techniques and theranostics, present innovative methods in recent years, enabling researchers and clinicians to overcome the limitations of unimodal imaging modalities and further advancing accuracy in the diagnosis and treatment of important pathologies, particularly cancer. Overall, nanoparticle-based imaging represents a transformative approach in advanced imaging modalities, offering new approaches to limitations of conventional agents currently being applied in clinical settings.
Collapse
Affiliation(s)
- Ahmad Butt
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| | - Horacio Bach
- Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
- Division of Infectious Diseases, Faculty of Medicine, The University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
3
|
Baniasadi A, Das JP, Prendergast CM, Beizavi Z, Ma HY, Jaber MY, Capaccione KM. Imaging at the nexus: how state of the art imaging techniques can enhance our understanding of cancer and fibrosis. J Transl Med 2024; 22:567. [PMID: 38872212 PMCID: PMC11177383 DOI: 10.1186/s12967-024-05379-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2024] [Accepted: 06/06/2024] [Indexed: 06/15/2024] Open
Abstract
Both cancer and fibrosis are diseases involving dysregulation of cell signaling pathways resulting in an altered cellular microenvironment which ultimately leads to progression of the condition. The two disease entities share common molecular pathophysiology and recent research has illuminated the how each promotes the other. Multiple imaging techniques have been developed to aid in the early and accurate diagnosis of each disease, and given the commonalities between the pathophysiology of the conditions, advances in imaging one disease have opened new avenues to study the other. Here, we detail the most up-to-date advances in imaging techniques for each disease and how they have crossed over to improve detection and monitoring of the other. We explore techniques in positron emission tomography (PET), magnetic resonance imaging (MRI), second generation harmonic Imaging (SGHI), ultrasound (US), radiomics, and artificial intelligence (AI). A new diagnostic imaging tool in PET/computed tomography (CT) is the use of radiolabeled fibroblast activation protein inhibitor (FAPI). SGHI uses high-frequency sound waves to penetrate deeper into the tissue, providing a more detailed view of the tumor microenvironment. Artificial intelligence with the aid of advanced deep learning (DL) algorithms has been highly effective in training computer systems to diagnose and classify neoplastic lesions in multiple organs. Ultimately, advancing imaging techniques in cancer and fibrosis can lead to significantly more timely and accurate diagnoses of both diseases resulting in better patient outcomes.
Collapse
Affiliation(s)
- Alireza Baniasadi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA.
| | - Jeeban P Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Conor M Prendergast
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Zahra Beizavi
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | - Hong Y Ma
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| | | | - Kathleen M Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 622 W 168Th Street, New York, NY, 10032, USA
| |
Collapse
|
4
|
Forma A, Grunwald A, Zembala P, Januszewski J, Brachet A, Zembala R, Świątek K, Baj J. Micronutrient Status and Breast Cancer: A Narrative Review. Int J Mol Sci 2024; 25:4968. [PMID: 38732186 PMCID: PMC11084730 DOI: 10.3390/ijms25094968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/28/2024] [Accepted: 04/30/2024] [Indexed: 05/13/2024] Open
Abstract
Breast cancer is one of the most common cancers worldwide, at the same time being one of the most prevalent causes of women's death. Many factors such as alcohol, weight fluctuations, or hormonal replacement therapy can potentially contribute to breast cancer development and progression. Another important factor in breast cancer onset includes micronutrient status. In this narrative review, we analyzed 23 micronutrients and their possible influence on breast cancer onset and progression. Further, the aim of this study was to investigate the impact of micronutrient status on the prevention of breast cancer and its possible influence on various therapeutic pathways. We researched meta-analyses, systemic and narrative reviews, retrospective studies, as well as original studies on human and animal models. The results of these studies indicate a possible correlation between the different levels of micronutrients and a decreased risk of breast cancer as well as a better survival rate. However, further studies are necessary to establish adequate doses of supplementation of the chosen micronutrients and the exact mechanisms of micronutrient impact on breast cancer therapy.
Collapse
Affiliation(s)
- Alicja Forma
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Arkadiusz Grunwald
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Patryk Zembala
- Faculty of Medicine, Medical University of Warsaw, Banacha 1A, 02-097 Warsaw, Poland;
| | - Jacek Januszewski
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| | - Adam Brachet
- Chair and Department of Forensic Medicine, Medical University of Lublin, Jaczewskiego 8b, 20-090 Lublin, Poland; (A.G.); (A.B.)
| | - Roksana Zembala
- Faculty of Medicine, Cardinal Stefan Wyszynski University in Warsaw, Wóycickiego 1/3, 01-938 Warsaw, Poland;
| | - Kamila Świątek
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| | - Jacek Baj
- Department of Correct, Clinical and Imaging Anatomy, Chair of Fundamental Sciences, Medical University of Lublin, Jaczewskiego 4, 20-090 Lublin, Poland; (J.J.); (K.Ś.); (J.B.)
| |
Collapse
|
5
|
Liu N, Wan Q, Wu X, Zhao T, Jakobsson V, Yuan H, Chen X, Zhang J, Zhang W. A comparison of [ 18F]AlF- and 68Ga-labeled dual targeting heterodimer FAPI-RGD in malignant tumor: preclinical evaluation and pilot clinical PET/CT imaging. Eur J Nucl Med Mol Imaging 2024; 51:1685-1697. [PMID: 38246909 DOI: 10.1007/s00259-023-06587-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Accepted: 12/12/2023] [Indexed: 01/23/2024]
Abstract
Due to the heterogeneity of tumors, strategies to improve the effectiveness of dual-targeting tracers in tumor diagnostics have been intensively practiced. In this study, the radiolabeled [18F]AlF-NOTA-FAPI-RGD (denoted as [18F]AlF-LNC1007), a dual-targeting heterodimer tracer targeting both fibroblast activation protein (FAP) and integrin αvβ3 to enhance specific tumor uptake and retention, was synthesized and evaluated. The tracer was compared with [68Ga]Ga-LNC1007 in preclinical and clinical settings. METHODS The preparation of [18F]AlF- and 68Ga-labeled FAPI-RGD was carried out with an optimized protocol. The stability was tested in PBS and fetal bovine serum (FBS). Cellular uptake and in vivo distribution of the two products were compared and carried out on the U87MG cell line and its xenograft model. The safety and dosimetry of [18F]AlF-LNC1007 PET/CT scan were evaluated in six patients with malignant tumors. RESULTS Two radiolabeling protocols of [18F]AlF-/[68Ga]Ga-LNC1007 were developed and optimized to give a high yield of tracers with good stability. In vivo microPET images showed that the two tracers exhibited comparable pharmacokinetic characteristics, with high tumor uptake and prolonged tumor retention. In vivo distribution data showed that the target-to-non-target ratios of [18F]AlF-LNC1007 were similar to[68Ga]Ga-LNC1007. A total of six patients underwent [18F]AlF-LNC1007 PET/CT evaluation while two had head-to-head [18F]FDG PET/CT scans. The total body effective dose was 9.94E-03 mSv/MBq. The biodistribution curve showed optimal normal organ uptake with high tumor uptake and long retention of up to 3h p.i., and notably, the tumor-to-background ratio increased over time. CONCLUSION We successfully prepared an [18F]AlF-LNC1007 dual-targeting PET probe with comparable performances as [68Ga]Ga-LNC1007. With prolonged tumor retention and tumor specificity, it produced good imaging quality in preclinical and clinical translational studies, indicating that [18F]AlF-LNC1007 is a promising non-invasive tracer for detecting tumors expressing FAP and/or integrin avβ3, with the prospect of clinical implementation.
Collapse
Affiliation(s)
- Nan Liu
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Qiang Wan
- Department of Nuclear Medicine, Affiliated Hospital of Southwest Medical University, Luzhou, 646000, China
| | - Xiaoming Wu
- College of Nuclear Science and Technology, Harbin Engineering University, Harbin, 150001, China
| | - Tianzhi Zhao
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Vivianne Jakobsson
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Hongmei Yuan
- Department of Pharmaceutics, School of Pharmacy, Southwest Medical University, Jiangyang District, Luzhou, 646000, China
| | - Xiaoyuan Chen
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
- Departments of Surgery, Chemical and Biomolecular Engineering, and Biomedical Engineering, Yong Loo Lin School of Medicine and College of Design and Engineering, National University of Singapore, Singapore, 119074, Singapore.
- Institute of Molecular and Cell Biology, Agency for Science, Technology, and Research (A*STAR), National University of Singapore, 61 Biopolis Drive, Proteos, Singapore, 138673, Singapore.
| | - Jingjing Zhang
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119074, Singapore.
- Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117599, Singapore.
- Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore.
| | - Wei Zhang
- Department of Nuclear Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China.
| |
Collapse
|
6
|
Xu Y, Chen J, Zhang Y, Zhang P. Recent Progress in Peptide-Based Molecular Probes for Disease Bioimaging. Biomacromolecules 2024; 25:2222-2242. [PMID: 38437161 DOI: 10.1021/acs.biomac.3c01413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/06/2024]
Abstract
Recent strides in molecular pathology have unveiled distinctive alterations at the molecular level throughout the onset and progression of diseases. Enhancing the in vivo visualization of these biomarkers is crucial for advancing disease classification, staging, and treatment strategies. Peptide-based molecular probes (PMPs) have emerged as versatile tools due to their exceptional ability to discern these molecular changes with unparalleled specificity and precision. In this Perspective, we first summarize the methodologies for crafting innovative functional peptides, emphasizing recent advancements in both peptide library technologies and computer-assisted peptide design approaches. Furthermore, we offer an overview of the latest advances in PMPs within the realm of biological imaging, showcasing their varied applications in diagnostic and therapeutic modalities. We also briefly address current challenges and potential future directions in this dynamic field.
Collapse
Affiliation(s)
- Ying Xu
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Junfan Chen
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| | - Yuan Zhang
- Department of Pulmonary and Critical Care Medicine, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, China
| | - Pengcheng Zhang
- School of Biomedical Engineering and State Key Laboratory of Advanced Medical Materials and Devices, ShanghaiTech University, Shanghai 201210, China
| |
Collapse
|
7
|
Ndlovu H, Lawal IO, Mokoala KMG, Sathekge MM. Imaging Molecular Targets and Metabolic Pathways in Breast Cancer for Improved Clinical Management: Current Practice and Future Perspectives. Int J Mol Sci 2024; 25:1575. [PMID: 38338854 PMCID: PMC10855575 DOI: 10.3390/ijms25031575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 01/23/2024] [Accepted: 01/24/2024] [Indexed: 02/12/2024] Open
Abstract
Breast cancer is the most frequently diagnosed cancer and leading cause of cancer-related deaths worldwide. Timely decision-making that enables implementation of the most appropriate therapy or therapies is essential for achieving the best clinical outcomes in breast cancer. While clinicopathologic characteristics and immunohistochemistry have traditionally been used in decision-making, these clinical and laboratory parameters may be difficult to ascertain or be equivocal due to tumor heterogeneity. Tumor heterogeneity is described as a phenomenon characterized by spatial or temporal phenotypic variations in tumor characteristics. Spatial variations occur within tumor lesions or between lesions at a single time point while temporal variations are seen as tumor lesions evolve with time. Due to limitations associated with immunohistochemistry (which requires invasive biopsies), whole-body molecular imaging tools such as standard-of-care [18F]FDG and [18F]FES PET/CT are indispensable in addressing this conundrum. Despite their proven utility, these standard-of-care imaging methods are often unable to image a myriad of other molecular pathways associated with breast cancer. This has stimulated interest in the development of novel radiopharmaceuticals targeting other molecular pathways and processes. In this review, we discuss validated and potential roles of these standard-of-care and novel molecular approaches. These approaches' relationships with patient clinicopathologic and immunohistochemical characteristics as well as their influence on patient management will be discussed in greater detail. This paper will also introduce and discuss the potential utility of novel PARP inhibitor-based radiopharmaceuticals as non-invasive biomarkers of PARP expression/upregulation.
Collapse
Affiliation(s)
- Honest Ndlovu
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa; (H.N.); (K.M.G.M.)
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
| | - Ismaheel O. Lawal
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA 30322, USA
| | - Kgomotso M. G. Mokoala
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa; (H.N.); (K.M.G.M.)
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
| | - Mike M. Sathekge
- Nuclear Medicine Research Infrastructure (NuMeRI), Steve Biko Academic Hospital, Pretoria 0001, South Africa; (H.N.); (K.M.G.M.)
- Department of Nuclear Medicine, University of Pretoria & Steve Biko Academic Hospital, Private Bag X169, Pretoria 0001, South Africa;
| |
Collapse
|
8
|
Vorster M, Hadebe BP, Sathekge MM. Theranostics in breast cancer. FRONTIERS IN NUCLEAR MEDICINE (LAUSANNE, SWITZERLAND) 2023; 3:1236565. [PMID: 39355052 PMCID: PMC11440857 DOI: 10.3389/fnume.2023.1236565] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 07/21/2023] [Indexed: 10/03/2024]
Abstract
Introduction Breast cancer is a complex disease and constitutes the leading cause of cancer in women globally. Conventional treatment modalities include surgery, chemotherapy, radiation therapy, and hormonal therapy; all of these have their limitations and often result in significant side effects or toxicity. Targeted radionuclide therapy based on a theranostic approach has been successfully applied in several malignancies, such as prostate cancer, thyroid cancer, and neuro-endocrine tumours. Several studies have also highlighted the potential of theranostic applications in breast cancer. Aim This review aims to provide an overview of the most promising current and future theranostic approaches in breast cancer. Discussion The discussion includes pre-clinical as well as clinical data on some of the most successful targets used to date. Examples of potential theranostic approaches include those targeting the Human epidermal growth factor receptor 2 (HER2) expression, angiogenesis, aspects of the tumour microenvironment, Gastrin-releasing peptide receptor (GRPR), Prostate-specific membrane antigen (PSMA) and Chemokine receptor 4 (CXCR-4) expression. Several challenges to widespread clinical implementation remain, which include regulatory approval, access to the various radiopharmaceuticals and imaging technology, cost-effectiveness, and the absence of robust clinical data. Conclusion Theranostic approaches have the potential to greatly improve diagnosis, treatment, and outcomes for patients with breast cancer. More research is needed to fully explore the potential of such approaches and to identify the best potential targets, considering feasibility, costs, efficacy, side effects and outcomes.
Collapse
Affiliation(s)
- M. Vorster
- Department of Nuclear Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - B. P. Hadebe
- Department of Nuclear Medicine, University of KwaZulu-Natal, Durban, South Africa
| | - M. M. Sathekge
- Department of Nuclear Medicine, University of Pretoria, Pretoria, South Africa
| |
Collapse
|
9
|
He D, Li H, Li Y, Xu Z, Wang C, Tang Y, Wu F, Zhen X, Wang S. Tumor-targeting semiconducting polymer nanoparticles: efficient adjuvant photothermal therapy using ultra-low laser power inhibits recurrences after breast-conserving surgery. NANOSCALE 2023; 15:6252-6262. [PMID: 36908261 DOI: 10.1039/d2nr06692k] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/18/2023]
Abstract
The need for adjuvant therapy to inhibit local recurrence after breast-conserving surgery with minimal side effects is great. Adjuvant photothermal therapy (aPTT) has the potential to replace radiotherapy and eliminates its inherent damage to healthy tissues. Herein, we functionalized semiconducting polymer nanoparticles (SPNs) with cRGD-peptide and silicon 2,3-naphthalocyanine bis(trihexylsilyloxide) (NIR775) to target breast cancer and perform aPTT under an ultra-low laser power (0.2 W cm-2) after breast-conserving surgery (BCS). The synthesized RGD-SPNNIR775 showed an excellent photothermal conversion efficiency and biocompatibility and was demonstrated to accumulate in tumors specifically. The BCS could be performed with confidence under the guidance of preoperative and postoperative fluorescence imaging. Notably, the aPTT completely inhibited the local recurrence after the BCS without compromising the cosmetic effect of the BCS. These results indicate the prospect of RGD-SPNNIR775 as a theranostic nanoplatform for efficient aPTT using an ultra-low laser power to control recurrence after BCS.
Collapse
Affiliation(s)
- Doudou He
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Haoze Li
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.
| | - Yang Li
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Ziqing Xu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Chuanbin Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Yuxia Tang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Feiyun Wu
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| | - Xu Zhen
- MOE Key Laboratory of High Performance Polymer Materials and Technology and Department of Polymer Science & Engineering, College of Chemistry & Chemical Engineering, Nanjing University, Nanjing, 210023, P. R. China.
| | - Shouju Wang
- Laboratory of Molecular Imaging, Department of Radiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, Jiangsu, China.
| |
Collapse
|
10
|
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
|
11
|
Hadebe B, Harry L, Ebrahim T, Pillay V, Vorster M. The Role of PET/CT in Breast Cancer. Diagnostics (Basel) 2023; 13:diagnostics13040597. [PMID: 36832085 PMCID: PMC9955497 DOI: 10.3390/diagnostics13040597] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/05/2023] [Accepted: 01/16/2023] [Indexed: 02/08/2023] Open
Abstract
Female breast cancer has surpassed lung cancer as the most commonly diagnosed cancer worldwide, with an estimated 2.3 million new cases (11.7%), followed by lung cancer (11.4%) The current literature and the National Comprehensive Cancer Network (NCCN) guidelines state that 18F-FDG PET/CT is not routine for early diagnosis of breast cancer, and rather PET/CT scanning should be performed for patients with stage III disease or when conventional staging studies yield non-diagnostic or suspicious results because this modality has been shown to upstage patients compared to conventional imaging and thus has an impact on disease management and prognosis. Furthermore, with the growing interest in precision therapy in breast cancer, numerous novel radiopharmaceuticals have been developed that target tumor biology and have the potential to non-invasively guide the most appropriate targeted therapy. This review discusses the role of 18F-FDG PET and other PET tracers beyond FDG in breast cancer imaging.
Collapse
Affiliation(s)
- Bawinile Hadebe
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
- Correspondence:
| | - Lerwine Harry
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Tasmeera Ebrahim
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Venesen Pillay
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| | - Mariza Vorster
- Department of Nuclear Medicine, College of Health Sciences, University of KwaZulu Natal, Private Bag X54001, Durban 4001, South Africa
- Inkosi Albert Luthuli Central Hospital, Durban 4001, South Africa
| |
Collapse
|
12
|
Carlsen EA, Loft M, Loft A, Czyzewska D, Andreassen M, Langer SW, Knigge U, Kjaer A. Prospective Phase II Trial of [ 68Ga]Ga-NODAGA-E[c(RGDyK)] 2 PET/CT Imaging of Integrin α vβ 3 for Prognostication in Patients with Neuroendocrine Neoplasms. J Nucl Med 2023; 64:252-259. [PMID: 35981899 PMCID: PMC9902862 DOI: 10.2967/jnumed.122.264383] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2022] [Revised: 08/13/2022] [Accepted: 08/13/2022] [Indexed: 02/04/2023] Open
Abstract
Integrin αvβ3, a subtype of the arginine-glycine-aspartate (RGD)-recognizing cell surface integrins, is upregulated on endothelial cells during angiogenesis and on tumor cells. Because of involvement in tumor growth, invasiveness and metastases, and angiogenesis, integrin αvβ3 is an attractive target in cancers. In this study, we applied 68Ga-NODAGA-E[c(RGDyK)]2 for imaging of integrin αvβ3 in patients with neuroendocrine neoplasms (NENs) and its potential use for prognostication. We hypothesized that 68Ga-NODAGA-E[c(RGDyK)]2 PET/CT would show tumor lesion uptake and that higher tumor lesion uptake was associated with a poorer prognosis. Methods: Between December 2017 and November 2020 we prospectively enrolled 113 patients with NEN of all grades (2019 World Health Organization classification) for 68Ga-NODAGA-E[c(RGDyK)]2 PET/CT. The scan was acquired 45 min after injection of 200 MBq of 68Ga-NODAGA-E[c(RGDyK)]2 Board-certified specialists in nuclear medicine and radiology analyzed the PET/CT measuring SUVmax in tumor lesions. Positive tumor lesions were defined as those with tumor-to-liver background ≥ 2. Maximal tumor SUVmax for each patient was used as a predictor of outcome. Patients were followed for at least 1 y to assess progression-free survival and overall survival. Results: Of 113 patients enrolled in the trial, 99 underwent 68Ga-NODAGA-E[c(RGDyK)]2 PET/CT, with 97 patients having evaluable lesions. The patients predominantly had small intestinal (64%) or pancreatic (20%) NEN and most had metastatic disease (93%). Most patients had low-grade tumors (78%), whereas 22% had high-grade tumors. During a median follow-up of 31 mo (interquartile range, 26-38 mo), 62 patients (64%) experienced disease progression and 25 (26%) patients died. In total, 76% of patients had positive tumor lesions, and of the patients with high-grade tumors 91% had positive tumor lesions. High integrin αvβ3 expression, defined as an SUVmax of at least 5.25, had a hazard ratio of 2.11 (95% CI, 1.18-3.78) and 6.95 (95% CI, 1.64-29.51) for progression-free survival and overall survival, respectively (P = 0.01 for both). Conclusion: Tumor lesion uptake of 68Ga-NODAGA-E[c(RGDyK)]2 was evident in patients with all grades of NEN. High uptake was associated with a poorer prognosis. Further studies are warranted to establish whether 68Ga-NODAGA-E[c(RGDyK)]2 PET/CT may become a prediction tool for identification of patients eligible for treatments targeting integrin αvβ3.
Collapse
Affiliation(s)
- Esben Andreas Carlsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital, Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Mathias Loft
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital, Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Annika Loft
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital, Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Dorota Czyzewska
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital, Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark;,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Mikkel Andreassen
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark;,Department of Clinical Endocrinology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Seppo W. Langer
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark;,Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark;,Department Clinical Medicine, University of Copenhagen, Copenhagen, Denmark; and
| | - Ulrich Knigge
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark;,Department of Clinical Endocrinology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark;,Department of Surgical Gastroenterology, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital, Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, Copenhagen, Denmark; .,ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark
| |
Collapse
|
13
|
de Mooij CM, Ploumen RAW, Nelemans PJ, Mottaghy FM, Smidt ML, van Nijnatten TJA. The influence of receptor expression and clinical subtypes on baseline [18F]FDG uptake in breast cancer: systematic review and meta-analysis. EJNMMI Res 2023; 13:5. [PMID: 36689007 PMCID: PMC9871105 DOI: 10.1186/s13550-023-00953-y] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/11/2023] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND To quantify the relationship between [18F]FDG uptake of the primary tumour measured by PET-imaging with immunohistochemical (IHC) expression of ER, PR, HER2, Ki-67, and clinical subtypes based on these markers in breast cancer patients. METHODS PubMed and Embase were searched for studies that compared SUVmax between breast cancer patients negative and positive for IHC expression of ER, PR, HER2, Ki-67, and clinical subtypes based on these markers. Two reviewers independently screened the studies and extracted the data. Standardized mean differences (SMD) and 95% confidence intervals (CIs) were estimated by using DerSimonian-Laird random-effects models. P values less than or equal to 5% indicated statistically significant results. RESULTS Fifty studies were included in the final analysis. SUVmax is significantly higher in ER-negative (31 studies, SMD 0.66, 0.56-0.77, P < 0.0001), PR-negative (30 studies, SMD 0.56; 0.40-0.71, P < 0.0001), HER2-positive (32 studies, SMD - 0.29, - 0.49 to - 0.10, P = 0.0043) or Ki-67-positive (19 studies, SMD - 0.77; - 0.93 to - 0.61, P < 0.0001) primary tumours compared to their counterparts. The majority of clinical subtypes were either luminal A (LA), luminal B (LB), HER2-positive or triple negative breast cancer (TNBC). LA is associated with significantly lower SUVmax compared to LB (11 studies, SMD - 0.49, - 0.68 to - 0.31, P = 0.0001), HER2-positive (15 studies, SMD - 0.91, - 1.21 to - 0.61, P < 0.0001) and TNBC (17 studies, SMD - 1.21, - 1.57 to - 0.85, P < 0.0001); and LB showed significantly lower uptake compared to TNBC (10 studies, SMD - 0.77, - 1.05 to - 0.49, P = 0.0002). Differences in SUVmax between LB and HER2-positive (9 studies, SMD - 0.32, - 0.88 to 0.24, P = 0.2244), and HER2-positive and TNBC (17 studies, SMD - 0.29, - 0.61 to 0.02, P = 0.0667) are not significant. CONCLUSION Primary tumour SUVmax is significantly higher in ER-negative, PR-negative, HER2-positive and Ki-67-positive breast cancer patients. Luminal tumours have the lowest and TNBC tumours the highest SUVmax. HER2 overexpression has an intermediate effect.
Collapse
Affiliation(s)
- Cornelis M de Mooij
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands.
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands.
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| | - Roxanne A W Ploumen
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Patty J Nelemans
- Department of Epidemiology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Felix M Mottaghy
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
- Department of Nuclear Medicine, University Hospital RWTH Aachen University, Aachen, Germany
| | - Marjolein L Smidt
- Department of Surgery, Maastricht University Medical Centre+, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Thiemo J A van Nijnatten
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Centre+, P.O. Box 5800, 6202 AZ, Maastricht, The Netherlands
- GROW - School for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
14
|
Pirasteh A, Periyasamy S, Meudt JJ, Liu Y, Lee LM, Schachtschneider KM, Schook LB, Gaba RC, Mao L, Said A, McMillan AB, Laeseke PF, Shanmuganayagam D. Staging Liver Fibrosis by Fibroblast Activation Protein Inhibitor PET in a Human-Sized Swine Model. J Nucl Med 2022; 63:1956-1961. [PMID: 35450958 PMCID: PMC9730920 DOI: 10.2967/jnumed.121.263736] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 04/07/2022] [Indexed: 01/11/2023] Open
Abstract
Current methods of staging liver fibrosis have notable limitations. We investigated the utility of PET in staging liver fibrosis by correlating liver uptake of 68Ga-labeled fibroblast activation protein inhibitor (FAPI) with histology in a human-sized swine model. Methods: Five pigs underwent baseline 68Ga-FAPI-46 (68Ga-FAPI) PET/MRI and liver biopsy, followed by liver parenchymal embolization, 8 wk of oral alcohol intake, endpoint 68Ga-FAPI PET/MRI, and necropsy. Regions of interest were drawn on baseline and endpoint PET images, and SUVmean was recorded. At the endpoint, liver sections corresponding to regions of interest were identified and cut out. Fibrosis was histologically evaluated using a modified METAVIR score for swine liver and quantitatively using collagen proportionate area (CPA). Box-and-whisker plots and linear regression were used to correlate SUVmean with METAVIR score and CPA, respectively. Results: Liver 68Ga-FAPI uptake strongly correlated with CPA (r = 0.89, P < 0.001). 68Ga-FAPI uptake was significantly and progressively higher across F2 and F3/F4 fibrosis stages, with a respective median SUVmean of 2.9 (interquartile range [IQR], 2.7-3.8) and 7.6 (IQR, 6.7-10.2) (P < 0.001). There was no significant difference between 68Ga-FAPI uptake of baseline liver and endpoint liver sections staged as F0/F1, with a respective median SUVmean of 1.7 (IQR, 1.3-2.0) and 1.7 (IQR, 1.5-1.8) (P = 0.338). Conclusion: The strong correlation between liver 68Ga-FAPI uptake and the histologic stage of liver fibrosis suggests that 68Ga-FAPI PET can play an impactful role in noninvasive staging of liver fibrosis, pending validation in patients.
Collapse
Affiliation(s)
- Ali Pirasteh
- Radiology and Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin
| | - Sarvesh Periyasamy
- Radiology and Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin
| | - Jennifer Jean Meudt
- Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, Wisconsin
| | - Yongjun Liu
- Pathology and Laboratory Medicine, University of Wisconsin–Madison, Madison, Wisconsin
| | - Laura M. Lee
- Research Animal Resources and Compliance, University of Wisconsin–Madison, Madison, Wisconsin
| | - Kyle M. Schachtschneider
- Radiology and Biochemistry and Molecular Genetics, University of Illinois at Chicago, Chicago, Illinois;,National Center for Supercomputing Applications, University of Illinois at Urbana–Champaign, Champaign, Illinois
| | - Lawrence B. Schook
- Animal Sciences, University of Illinois at Chicago, Chicago, Illinois;,Radiology/Interventional Radiology, University of Illinois at Chicago, Chicago, Illinois
| | - Ron C. Gaba
- Radiology/Interventional Radiology, University of Illinois at Chicago, Chicago, Illinois
| | - Lu Mao
- Biostatistics and Medical Informatics, University of Wisconsin–Madison, Madison, Wisconsin
| | - Adnan Said
- Medicine, Gastroenterology, and Hepatology, University of Wisconsin–Madison, Madison, Wisconsin;,William S. Middleton VA Medical Center, Madison, Wisconsin
| | - Alan Blair McMillan
- Radiology and Medical Physics, University of Wisconsin–Madison, Madison, Wisconsin
| | - Paul F. Laeseke
- Radiology and Biomedical Engineering, University of Wisconsin–Madison, Madison, Wisconsin
| | - Dhanansayan Shanmuganayagam
- Animal and Dairy Sciences, University of Wisconsin–Madison, Madison, Wisconsin;,Surgery, University of Wisconsin–Madison, Madison, Wisconsin; and,Center for Biomedical Swine Research and Innovation, University of Wisconsin–Madison, Madison, Wisconsin
| |
Collapse
|
15
|
Wu J, Zhang X, Jia Z, Zhou X, Qi R, Ji H, Sun J, Sun C, Teng Z, Lu G, Chen X. Combined 18F-FDG and 18F-Alfatide II PET May Predict Luminal B (HER2 Negative) Subtype and Nonluminal Subtype of Invasive Breast Cancer. Mol Pharm 2022; 19:3405-3411. [PMID: 35972444 DOI: 10.1021/acs.molpharmaceut.2c00547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Noninvasive PET molecular imaging using radiopharmaceuticals is important to classify breast cancer in the clinic. The aim of this study was to investigate the combination of 18F-FDG and 18F-Alfatide II for predicting molecular subtypes of invasive breast cancer. Forty-four female patients with clinically suspected breast cancer were recruited and underwent 18F-FDG and 18F-Alfatide II PET/CT within a week. Tracer uptake in breast lesions was assessed using the maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), and SUVmax ratio of 18F-FDG to 18F-Alfatide II (FAR). Invasive breast cancer lesions were further classified as luminal A subtype, luminal B subtype, human epidermal growth factor receptor-2 (HER2) overexpressing subtype, and triple negative subtype according to the expression of the estrogen receptor (ER), progesterone receptor (PR), HER2, and Ki-67. Among 44 patients, 35 patients were pathologically diagnosed with invasive breast cancer. The SUVmax and SUVmean of 18F-FDG were significantly higher in the ER-negative group than those in the ER-positive group, as well as in the PR-negative group than those in the PR-positive group. However, the SUVmax and SUVmean of 18F-Alfatide II were higher in the ER-positive group and the PR-positive group. By combining 18F-FDG and 18F-Alfatide II, the FAR was lower in the ER-positive group and the PR-positive group. The HER2 overexpressing subtype showed the highest SUVmax and SUVmean for 18F-FDG while the luminal B (HER2 negative) subtype revealed the lowest values. The luminal B (HER2 negative) subtype showed the highest 18F-Alfatide II SUVmax, while the triple negative subtype showed the lowest 18F-Alfatide II SUVmax. The FAR was the lowest in the luminal B (HER2 negative) subtype and much higher in the HER2 overexpressing and triple negative subtypes. FAR less than 1 predicted the luminal B (HER2 negative) subtype with high specificity (93.1%) and NPV (90%). FAR greater than 3 predicted the HER2 overexpressing subtype and triple negative subtype (namely, the nonluminal subtype) with very high specificity (100%) and PPV (100%). In summary, FAR, the combined PET parameter of 18F-FDG and 18F-Alfatide II, can be used to predict molecular subtypes of invasive breast cancer, especially for the luminal B (HER2 negative) subtype and the nonluminal subtype.
Collapse
Affiliation(s)
- Jiang Wu
- Department of Nuclear Medicine, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Xiaoyi Zhang
- Department of Nuclear Medicine, Changshu No.2 People's Hospital, Changshu 215500, China
| | - Zhijun Jia
- Department of Nuclear Medicine, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing 210008, China
| | - Xiaodie Zhou
- Department of Pathology, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Rongxin Qi
- Department of Nuclear Medicine, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Hengshan Ji
- Department of Nuclear Medicine, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Jingjing Sun
- Department of Nuclear Medicine, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Chuanjin Sun
- Department of Nuclear Medicine, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Zhaogang Teng
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials, Jiangsu National Synergetic Innovation Centre for Advanced Materials, Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Guangming Lu
- Department of Diagnostic Radiology, Jinling Hospital, Medical School, Nanjing University, Nanjing 210002, China
| | - Xiaoyuan Chen
- Departments of Diagnostic Radiology, and Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Departments of Chemical and Biomolecular Engineering, and Biomedical Engineering, National University of Singapore, Singapore 117599, Singapore.,Clinical Imaging Research Centre, Centre for Translational Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.,Nanomedicine Translational Research Program, NUS Center for Nanomedicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
16
|
Lv R, Raab M, Wang Y, Tian J, Lin J, Prasad PN. Nanochemistry advancing photon conversion in rare-earth nanostructures for theranostics. Coord Chem Rev 2022. [DOI: 10.1016/j.ccr.2022.214486] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Ebrahimi F, Hosseinimehr SJ. Homomultimer strategy for improvement of radiolabeled peptides and antibody fragments in tumor targeting. Curr Med Chem 2022; 29:4923-4957. [PMID: 35450521 DOI: 10.2174/0929867329666220420131836] [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: 11/23/2021] [Revised: 01/18/2022] [Accepted: 02/07/2022] [Indexed: 11/22/2022]
Abstract
A homomultimeric radioligand is composed of multiple identical ligands connected to the linker and radionuclide to detect a variety of overexpressed receptors on cancer cells. Multimer strategy holds great potential for introducing new radiotracers based on peptide and monoclonal antibody (mAb) derivatives in molecular imaging and therapy. It offers a reliable procedure for the preparation of biological-based targeting with diverse affinities and pharmacokinetics. In this context, we provide a useful summary and interpretation of the main results by a comprehensive look at multimeric radiopharmaceuticals in nuclear oncology. Therefore, there will be explanations for the strategy mechanisms and the main variables affecting the biodistribution results. The discussion is followed by highlights of recent work in the targeting of various types of receptors. The consequences are expressed based on comparing some parameters between monomer and multimer counterparts in each relevant section.
Collapse
Affiliation(s)
- Fatemeh Ebrahimi
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Seyed Jalal Hosseinimehr
- Department of Radiopharmacy, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
18
|
Clausen MM, Carlsen EA, Christensen C, Madsen J, Brandt-Larsen M, Klausen TL, Holm S, Loft A, Berthelsen AK, Kroman N, Knigge U, Kjaer A. First-in-Human Study of [68Ga]Ga-NODAGA-E[c(RGDyK)]2 PET for Integrin αvβ3 Imaging in Patients with Breast Cancer and Neuroendocrine Neoplasms: Safety, Dosimetry and Tumor Imaging Ability. Diagnostics (Basel) 2022; 12:diagnostics12040851. [PMID: 35453899 PMCID: PMC9027224 DOI: 10.3390/diagnostics12040851] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 03/28/2022] [Indexed: 02/01/2023] Open
Abstract
Arginine-Glycine-Aspartate (RGD)-recognizing cell surface integrins are involved in tumor growth, invasiveness/metastases, and angiogenesis, and are therefore an attractive treatment target in cancers. The subtype integrin αvβ3 is upregulated on endothelial cells during angiogenesis and on tumor cells. In vivo assessment of integrin αvβ3 is possible with positron emission tomography (PET). Preclinical data on radiochemical properties, tumor uptake and radiation exposure identified [68Ga]Ga-NODAGA-E[c(RGDyK)]2 as a promising candidate for clinical translation. In this first-in-human phase I study, we evaluate [68Ga]Ga-NODAGA-E[c(RGDyK)]2 PET in patients with neuroendocrine neoplasms (NEN) and breast cancer (BC). The aim was to investigate safety, biodistribution and dosimetry as well as tracer uptake in tumor lesions. A total of 10 patients (5 breast cancer, 5 neuroendocrine neoplasm) received a single intravenous dose of approximately 200 MBq [68Ga]Ga-NODAGA-E[c(RGDyK)]2. Biodistribution profile and dosimetry were assessed by whole-body PET/CT performed at 10 min, 1 h and 2 h after injection. Safety assessment with vital parameters, electrocardiograms and blood tests were performed before and after injection. In vivo stability of [68Ga]Ga-NODAGA-E[c(RGDyK)]2 was determined by analysis of blood and urine. PET images were analyzed for tracer uptake in tumors and background organs. No adverse events or pharmacologic effects were observed in the 10 patients. [68Ga]Ga-NODAGA-E[c(RGDyK)]2 exhibited good in vivo stability and fast clearance, primarily by renal excretion. The effective dose was 0.022 mSv/MBq, equaling a radiation exposure of 4.4 mSv at an injected activity of 200 MBq. The tracer demonstrated stable tumor retention and good image contrast. In conclusion, this first-in-human phase I trial demonstrated safe use of [68Ga]Ga-NODAGA-E[c(RGDyK)]2 for integrin αvβ3 imaging in cancer patients, low radiation exposure and favorable uptake in tumors. Further studies are warranted to establish whether [68Ga]Ga-NODAGA-E[c(RGDyK)]2 may become a tool for early identification of patients eligible for treatments targeting integrin αvβ3 and for risk stratification of patients.
Collapse
Affiliation(s)
- Malene Martini Clausen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- Department of Oncology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Esben Andreas Carlsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Camilla Christensen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Jacob Madsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Malene Brandt-Larsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Thomas Levin Klausen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Søren Holm
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Annika Loft
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Anne Kiil Berthelsen
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
| | - Niels Kroman
- Department of Breast Surgery, Copenhagen University Hospital—Herlev/Gentofte Hospital, 2730 Herlev, Denmark;
| | - Ulrich Knigge
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
- Departments of Clinical Endocrinology and Surgical Gastroenterology, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark
| | - Andreas Kjaer
- Department of Clinical Physiology and Nuclear Medicine & Cluster for Molecular Imaging, Copenhagen University Hospital—Rigshospitalet & Department of Biomedical Sciences, University of Copenhagen, 2100 Copenhagen, Denmark; (M.M.C.); (E.A.C.); (C.C.); (J.M.); (M.B.-L.); (T.L.K.); (S.H.); (A.L.); (A.K.B.)
- ENETS Neuroendocrine Tumor Center of Excellence, Copenhagen University Hospital—Rigshospitalet, 2100 Copenhagen, Denmark;
- Correspondence:
| |
Collapse
|
19
|
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
|
20
|
Li Q, Lan T, Xie J, Lu Y, Zheng D, Su B. Integrin-Mediated Tumorigenesis and Its Therapeutic Applications. Front Oncol 2022; 12:812480. [PMID: 35223494 PMCID: PMC8873568 DOI: 10.3389/fonc.2022.812480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/17/2022] [Indexed: 11/13/2022] Open
Abstract
Integrins, a family of adhesion molecules generally exist on the cell surface, are essential for regulating cell growth and its function. As a bi-directional signaling molecule, they mediate cell-cell and cell-extracellular matrix interaction. The recognitions of their key roles in many human pathologies, including autoimmunity, thrombosis and neoplasia, have revealed their great potential as a therapeutic target. This paper focuses on the activation of integrins, the role of integrins in tumorigenesis and progression, and advances of integrin-dependent tumor therapeutics in recent years. It is expected that understanding function and signaling transmission will fully exploit potentialities of integrin as a novel target for tumors.
Collapse
Affiliation(s)
- Qingling Li
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ting Lan
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Jian Xie
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Youguang Lu
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Dali Zheng
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- *Correspondence: Dali Zheng, ; Bohua Su,
| | - Bohua Su
- Fujian Key Laboratory of Oral Diseases, Fujian Provincial Engineering Research Center of Oral Biomaterial, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Preventive Dentistry, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- *Correspondence: Dali Zheng, ; Bohua Su,
| |
Collapse
|
21
|
18F-Alfatide II for the evaluation of axillary lymph nodes in breast cancer patients: comparison with 18F-FDG. Eur J Nucl Med Mol Imaging 2022; 49:2869-2876. [PMID: 35138445 DOI: 10.1007/s00259-021-05333-z] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Accepted: 03/23/2021] [Indexed: 11/04/2022]
Abstract
PURPOSE 18F-Alfatide II has been translated into clinical use and been proven to have good performance in identifying breast cancer. In this study, we investigated 18F-Alfatide II for evaluation of axillary lymph nodes (ALN) in breast cancer patients and compared the performance with 18F-FDG. METHODS A total of 44 female patients with clinically suspected breast cancer were enrolled and underwent 18F-Alfatide II and 18F-FDG PET/CT within a week. Tracer uptakes in ALN were evaluated by visual analysis, semi-quantitative analysis with maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), and SUVmax ratio of target/non-target (T/NT). RESULTS Among 44 patients, 37 patients were pathologically diagnosed with breast cancer with metastatic (17 cases) or non-metastatic (20 cases) ALN. The sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV) of visual analysis were 70.6%, 90%, 81.1%, 85.7%, and 78.3% for 18F-Alfatide II, 64.7%, 90%, 78.4%, 84.6%, and 75% for 18F-FDG, respectively. By combining 18F-Alfatide II and 18F-FDG, the sensitivity significantly increased to 82.4%, the specificity was 85%, the accuracy increased to 83.8%, the PPV was 82.4%, and the NPV significantly increased to 85.0%. Three cases of luminal B subtype were false negative for both 18F-Alfatide II and 18F-FDG. The other 2 false negative cases of 18F-Alfatide II were triple-negative subtype and 3 false negative cases of 18F-FDG were luminal B subtype too. The AUCs of three semi-quantitative parameters (SUVmax, SUVmean, T/NT) for 18F-Alfatide II were between 0.8 and 0.9, whereas those for 18F-FDG were more than 0.9. 18F-Alfatide II T/NT had the highest Youden index (76.5%), specificity (100%), accuracy (89.2%), and PPV (100%) among these semi-quantitative parameters. 18F-Alfatide II uptake as well as 18F-FDG uptake in metastatic axillary lymph nodes (MALN) was significantly higher than that in benign axillary lymph nodes (BALN). Both 18F-Alfatide II and 18F-FDG did not show difference in primary tumor uptake irrespective of ALN status. CONCLUSION 18F-Alfatide II can be used in breast cancer patients to detect metastatic ALN, however, like 18F-FDG, with high specificity but relatively low sensitivity. The combination of 18F-Alfatide II and 18F-FDG can significantly improve sensitivity and NPV. 18F-Alfatide II T/NT may serve as the most important semi-quantitative parameter to evaluate ALN.
Collapse
|
22
|
Han Z, Ke M, Liu X, Wang J, Guan Z, Qiao L, Wu Z, Sun Y, Sun X. Molecular Imaging, How Close to Clinical Precision Medicine in Lung, Brain, Prostate and Breast Cancers. Mol Imaging Biol 2022; 24:8-22. [PMID: 34269972 DOI: 10.1007/s11307-021-01631-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 07/03/2021] [Accepted: 07/06/2021] [Indexed: 12/15/2022]
Abstract
Precision medicine is playing a pivotal role in strategies of cancer therapy. Unlike conventional one-size-fits-all chemotherapy or radiotherapy modalities, precision medicine could customize an individual treatment plan for cancer patients to acquire superior efficacy, while minimizing side effects. Precision medicine in cancer therapy relies on precise and timely tumor biological information. Traditional tissue biopsies, however, are often inadequate in meeting this requirement due to cancer heterogeneity, poor tolerance, and invasiveness. Molecular imaging could detect tumor biology characterization in a noninvasive and visual manner, and provide information about therapeutic targets, treatment response, and pharmacodynamic evaluation. This summates to significant value in guiding cancer precision medicine in aspects of patient screening, treatment monitoring, and estimating prognoses. Although growing clinical evidences support the further application of molecular imaging in precision medicine of cancer, some challenges remain. In this review, we briefly summarize and discuss representative clinical trials of molecular imaging in improving precision medicine of cancer patients, aiming to provide useful references for facilitating further clinical translation of molecular imaging to precision medicine of cancers.
Collapse
Affiliation(s)
- Zhaoguo Han
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
- Biomedical Research Imaging Center, Department of Radiology, University of North Carolina-Chapel Hill, Chapel Hill, NC, USA
| | - Mingxing Ke
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xiang Liu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Jing Wang
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Zhengqi Guan
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Lina Qiao
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Zhexi Wu
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Yingying Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China
| | - Xilin Sun
- NHC and CAMS Key Laboratory of Molecular Probe and Targeted Theranostics, Molecular Imaging Research Center (MIRC), Harbin Medical University, 766 Xiangan N street, Harbin, 150028, Heilongjiang, China.
- Department of Nuclear Medicine, The Fourth Hospital of Harbin Medical University, Harbin, China.
| |
Collapse
|
23
|
Kossatz S, Beer AJ, Notni J. It's Time to Shift the Paradigm: Translation and Clinical Application of Non-αvβ3 Integrin Targeting Radiopharmaceuticals. Cancers (Basel) 2021; 13:cancers13235958. [PMID: 34885066 PMCID: PMC8657165 DOI: 10.3390/cancers13235958] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/16/2021] [Accepted: 11/17/2021] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Cancer cells often present a different set of proteins on their surface than normal cells. This also applies to integrins, a class of 24 cell surface receptors which mainly are responsible for physically anchoring cells in tissues, but also fulfil a plethora of other functions. If a certain integrin is found on tumor cells but not on normal ones, radioactive molecules (named tracers) that specifically bind to this integrin will accumulate in the cancer lesion if injected into the blood stream. The emitted radiation can be detected from outside the body and allows for localization and thus, diagnosis, of cancer. Only one of the 24 integrins, the subtype αvβ3, has hitherto been thoroughly investigated in this context. We herein summarize the most recent, pertinent research on other integrins, and argue that some of these approaches might ultimately improve the clinical management of the most lethal cancers, such as pancreatic carcinoma. Abstract For almost the entire period of the last two decades, translational research in the area of integrin-targeting radiopharmaceuticals was strongly focused on the subtype αvβ3, owing to its expression on endothelial cells and its well-established role as a biomarker for, and promoter of, angiogenesis. Despite a large number of translated tracers and clinical studies, a clinical value of αvβ3-integrin imaging could not be defined yet. The focus of research has, thus, been moving slowly but steadily towards other integrin subtypes which are involved in a large variety of tumorigenic pathways. Peptidic and non-peptidic radioligands for the integrins α5β1, αvβ6, αvβ8, α6β1, α6β4, α3β1, α4β1, and αMβ2 were first synthesized and characterized preclinically. Some of these compounds, targeting the subtypes αvβ6, αvβ8, and α6β1/β4, were subsequently translated into humans during the last few years. αvβ6-Integrin has arguably attracted most attention because it is expressed by some of the cancers with the worst prognosis (above all, pancreatic ductal adenocarcinoma), which substantiates a clinical need for the respective theranostic agents. The receptor furthermore represents a biomarker for malignancy and invasiveness of carcinomas, as well as for fibrotic diseases, such as idiopathic pulmonary fibrosis (IPF), and probably even for Sars-CoV-2 (COVID-19) related syndromes. Accordingly, the largest number of recent first-in-human applications has been reported for radiolabeled compounds targeting αvβ6-integrin. The results indicate a substantial clinical value, which might lead to a paradigm change and trigger the replacement of αvβ3 by αvβ6 as the most popular integrin in theranostics.
Collapse
Affiliation(s)
- Susanne Kossatz
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, 81675 Munich, Germany;
- Central Institute for Translational Cancer Research (TranslaTUM), School of Medicine, Technical University of Munich, 81675 Munich, Germany
| | | | - Johannes Notni
- Department of Pathology, School of Medicine, Technical University of Munich, 81675 Munich, Germany
- TRIMT GmbH, 01454 Radeberg, Germany
- Correspondence: ; Tel.: +49-89-4140-6075; Fax: +49-89-4140-6949
| |
Collapse
|
24
|
Steiger K, Quigley NG, Groll T, Richter F, Zierke MA, Beer AJ, Weichert W, Schwaiger M, Kossatz S, Notni J. There is a world beyond αvβ3-integrin: Multimeric ligands for imaging of the integrin subtypes αvβ6, αvβ8, αvβ3, and α5β1 by positron emission tomography. EJNMMI Res 2021; 11:106. [PMID: 34636990 PMCID: PMC8506476 DOI: 10.1186/s13550-021-00842-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND In the context of nuclear medicine and theranostics, integrin-related research and development was, for most of the time, focused predominantly on 'RGD peptides' and the subtype αvβ3-integrin. However, there are no less than 24 known integrins, and peptides without the RGD sequence as well as non-peptidic ligands play an equally important role as selective integrin ligands. On the other hand, multimerization is a well-established method to increase the avidity of binding structures, but multimeric radiopharmaceuticals have not made their way into clinics yet. In this review, we describe how these aspects have been interwoven in the framework of the German Research Foundation's multi-group interdisciplinary funding scheme CRC 824, yielding a series of potent PET imaging agents for selective imaging of various integrin subtypes. RESULTS The gallium-68 chelator TRAP was utilized to elaborate symmetrical trimers of various peptidic and non-peptidic integrin ligands. Preclinical data suggested a high potential of the resulting Ga-68-tracers for PET-imaging of the integrins α5β1, αvβ8, αvβ6, and αvβ3. For the first three, we provide some additional immunohistochemistry data in human cancers, which suggest several future clinical applications. Finally, application of αvβ3- and αvβ6-integrin tracers in pancreatic carcinoma patients revealed that unlike αvβ3-targeted PET, αvβ6-integrin PET is not characterized by off-target uptake and thus, enables a substantially improved imaging of this type of cancer. CONCLUSIONS Novel radiopharmaceuticals targeting a number of different integrins, above all, αvβ6, have proven their clinical potential and will play an increasingly important role in future theranostics.
Collapse
Affiliation(s)
- Katja Steiger
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Neil Gerard Quigley
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Tanja Groll
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Frauke Richter
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | | | | | - Wilko Weichert
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany
| | - Markus Schwaiger
- Klinik Für Nuklearmedizin Und Zentralinstitut Für Translationale Krebsforschung (TranslaTUM), Klinikum Rechts Der Isar der Technischen Universität München, Munich, Germany
| | - Susanne Kossatz
- Klinik Für Nuklearmedizin Und Zentralinstitut Für Translationale Krebsforschung (TranslaTUM), Klinikum Rechts Der Isar der Technischen Universität München, Munich, Germany
| | - Johannes Notni
- Institut Für Pathologie Und Pathologische Anatomie, Technische Universität München, Munich, Germany. .,Experimental Radiopharmacy, Clinic for Nuclear Medicine, University Hospital Essen, Hufelandstr. 55, 45147, Essen, Germany.
| |
Collapse
|
25
|
Lilburn DM, Groves AM. The role of PET in imaging of the tumour microenvironment and response to immunotherapy. Clin Radiol 2021; 76:784.e1-784.e15. [DOI: 10.1016/j.crad.2021.08.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
26
|
Archibald SJ, Allott L. The aluminium-[ 18F]fluoride revolution: simple radiochemistry with a big impact for radiolabelled biomolecules. EJNMMI Radiopharm Chem 2021; 6:30. [PMID: 34436693 PMCID: PMC8390636 DOI: 10.1186/s41181-021-00141-0] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 07/22/2021] [Indexed: 12/15/2022] Open
Abstract
The aluminium-[18F]fluoride ([18F]AlF) radiolabelling method combines the favourable decay characteristics of fluorine-18 with the convenience and familiarity of metal-based radiochemistry and has been used to parallel gallium-68 radiopharmaceutical developments. As such, the [18F]AlF method is popular and widely implemented in the development of radiopharmaceuticals for the clinic. In this review, we capture the current status of [18F]AlF-based technology and reflect upon its impact on nuclear medicine, as well as offering our perspective on what the future holds for this unique radiolabelling method.
Collapse
Affiliation(s)
- Stephen J Archibald
- Positron Emission Tomography Research Centre, Faculty of Health Sciences, University of Hull, Cottingham Road, Kingston upon Hull, HU6 7RX, UK.,Department of Biomedical Sciences, Faculty of Health Sciences, University of Hull, Cottingham Road, Kingston upon Hull, HU6 7RX, UK.,Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Castle Road, Cottingham, HU16 5JQ, UK
| | - Louis Allott
- Positron Emission Tomography Research Centre, Faculty of Health Sciences, University of Hull, Cottingham Road, Kingston upon Hull, HU6 7RX, UK. .,Department of Biomedical Sciences, Faculty of Health Sciences, University of Hull, Cottingham Road, Kingston upon Hull, HU6 7RX, UK. .,Hull University Teaching Hospitals NHS Trust, Castle Hill Hospital, Castle Road, Cottingham, HU16 5JQ, UK.
| |
Collapse
|
27
|
Yuan G, Liu S, Ma H, Su S, Wen F, Tang X, Zhang Z, Zhao J, Lin L, Xiang X, Nie D, Tang G. Targeting Phosphatidylethanolamine with Fluorine-18 Labeled Small Molecule Probe for Apoptosis Imaging. Mol Imaging Biol 2021; 22:914-923. [PMID: 31828718 DOI: 10.1007/s11307-019-01460-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
PURPOSE Externalization of phosphatidylethanolamine (PE) in dying cells makes the phospholipid an attractive target for apoptosis imaging. However, no ideal PE-targeted positron emission tomography (PET) radiotracer was developed. The goal of the study was to develop a novel PE-targeted radiopharmaceutical to imaging apoptosis. PROCEDURE In this study, we have radiolabeled PE-binding polypeptide duramycin with fluorine-18 for PET imaging of apoptosis. Al[18F]F-NOTA-PEG3-duramycin was synthesized via chelation reaction of NOTA-PEG3-duramycin with Al[18F]F. PE-binding capacity of Al[18F]F-NOTA-PEG3-duramycin was determined in a competitive radiometric PE-binding assay. The pharmacokinetic profile was evaluated in Kunming mice. The apoptosis imaging capacity of Al[18F]F-NOTA-PEG3-duramycin was evaluated using in vitro cell uptake assay with camptothecin-treated Jurkat cells, along with in vivo PET imaging using erlotinib-treated nude mice. RESULTS The total synthesis procedure lasted for 30 min, with a decay-uncorrected radiochemical yield of 21.3 ± 2.6 % (n = 10). Compared with the control cells, the binding of Al[18F]F-NOTA-PEG3-duramycin with camptothecin-induced apoptotic cells resulted in a tripling increase. A competitive radiometric PE-binding assay strongly confirmed the binding of Al[18F]F-NOTA-PEG3-duramycin to PE. The biodistribution study showed rapid blood clearance, prominent kidney retention, and low liver uptake. In the in vivo PET/CT imaging, Al[18F]F-NOTA-PEG3-duramycin demonstrated 2-fold increase in erlotinib-treated HCC827 tumors in nude mice. CONCLUSION Considering the facile preparation and improved biological properties, Al[18F]F-NOTA-PEG3-duramycin seems to be a promising PET tracer candidate for imaging apoptosis in the monitoring of cancer treatment.
Collapse
Affiliation(s)
- Gongjun Yuan
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shaoyu Liu
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Hui Ma
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Shu Su
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Fuhua Wen
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xiaolan Tang
- Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China.,School of Materials and Energy, South China Agricultural University, Guangzhou, 510642, China
| | - Zhanwen Zhang
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Jing Zhao
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Liping Lin
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Xianhong Xiang
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China
| | - Dahong Nie
- Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China. .,Department of Radiation Oncology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China.
| | - Ganghua Tang
- Department of Nuclear Medicine and Medical Imaging, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510080, China. .,Guangdong Engineering Research center for Translational Application of Medical Radiopharmaceuticals, Sun Yat-sen University, Guangzhou, 510080, China. .,Nanfang PET Center and Department of Nuclear Medicine, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
28
|
Staszak K, Wieszczycka K, Bajek A, Staszak M, Tylkowski B, Roszkowski K. Achievement in active agent structures as a power tools in tumor angiogenesis imaging. Biochim Biophys Acta Rev Cancer 2021; 1876:188560. [PMID: 33965512 DOI: 10.1016/j.bbcan.2021.188560] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/13/2021] [Accepted: 04/29/2021] [Indexed: 12/26/2022]
Abstract
According to World Health Organization (WHO) cancer is the second most important cause of death globally. Because angiogenesis is considered as an essential process of growth, proliferation and tumor progression, within this review we decided to shade light on recent development of chemical compounds which play a significant role in its imaging and monitoring. Indeed, the review gives insight about the current achievements of active agents structures involved in imaging techniques such as: positron emission computed tomography (PET), magnetic resonance imaging (MRI) and single photon emission computed tomography (SPECT), as well as combination PET/MRI and PET/CT. The review aims to provide the journal audience with a comprehensive and in-deep understanding of chemistry policy in tumor angiogenesis imaging.
Collapse
Affiliation(s)
- Katarzyna Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, 60-965 Poznan, Poland
| | - Karolina Wieszczycka
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, 60-965 Poznan, Poland
| | - Anna Bajek
- Department of Tissue Engineering, Collegium Medicum Nicolaus Copernicus University, Karlowicza St. 24, 85-092 Bydgoszcz, Poland
| | - Maciej Staszak
- Institute of Technology and Chemical Engineering, Poznan University of Technology, ul. Berdychowo 4, 60-965 Poznan, Poland
| | - Bartosz Tylkowski
- Eurecat, Centre Tecnològic de Catalunya, C/Marcellí Domingo s/n, 43007 Tarragona, Spain
| | - Krzysztof Roszkowski
- Department of Oncology, Collegium Medicum Nicolaus Copernicus University, Romanowskiej St. 2, 85-796 Bydgoszcz, Poland.
| |
Collapse
|
29
|
Ludwig BS, Kessler H, Kossatz S, Reuning U. RGD-Binding Integrins Revisited: How Recently Discovered Functions and Novel Synthetic Ligands (Re-)Shape an Ever-Evolving Field. Cancers (Basel) 2021; 13:1711. [PMID: 33916607 PMCID: PMC8038522 DOI: 10.3390/cancers13071711] [Citation(s) in RCA: 131] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 03/22/2021] [Accepted: 03/29/2021] [Indexed: 12/19/2022] Open
Abstract
Integrins have been extensively investigated as therapeutic targets over the last decades, which has been inspired by their multiple functions in cancer progression, metastasis, and angiogenesis as well as a continuously expanding number of other diseases, e.g., sepsis, fibrosis, and viral infections, possibly also Severe Acute Respiratory Syndrome Coronavirus (SARS-CoV-2). Although integrin-targeted (cancer) therapy trials did not meet the high expectations yet, integrins are still valid and promising targets due to their elevated expression and surface accessibility on diseased cells. Thus, for the future successful clinical translation of integrin-targeted compounds, revisited and innovative treatment strategies have to be explored based on accumulated knowledge of integrin biology. For this, refined approaches are demanded aiming at alternative and improved preclinical models, optimized selectivity and pharmacological properties of integrin ligands, as well as more sophisticated treatment protocols considering dose fine-tuning of compounds. Moreover, integrin ligands exert high accuracy in disease monitoring as diagnostic molecular imaging tools, enabling patient selection for individualized integrin-targeted therapy. The present review comprehensively analyzes the state-of-the-art knowledge on the roles of RGD-binding integrin subtypes in cancer and non-cancerous diseases and outlines the latest achievements in the design and development of synthetic ligands and their application in biomedical, translational, and molecular imaging approaches. Indeed, substantial progress has already been made, including advanced ligand designs, numerous elaborated pre-clinical and first-in-human studies, while the discovery of novel applications for integrin ligands remains to be explored.
Collapse
Affiliation(s)
- Beatrice S. Ludwig
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Susanne Kossatz
- Department of Nuclear Medicine, University Hospital Klinikum Rechts der Isar and Central Institute for Translational Cancer Research (TranslaTUM), Technical University Munich, 81675 Munich, Germany;
- Department of Chemistry, Institute for Advanced Study, Technical University Munich, 85748 Garching, Germany;
| | - Ute Reuning
- Clinical Research Unit, Department of Obstetrics and Gynecology, University Hospital Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
| |
Collapse
|
30
|
Liolios C, Sachpekidis C, Kolocouris A, Dimitrakopoulou-Strauss A, Bouziotis P. PET Diagnostic Molecules Utilizing Multimeric Cyclic RGD Peptide Analogs for Imaging Integrin α vβ 3 Receptors. Molecules 2021; 26:molecules26061792. [PMID: 33810198 PMCID: PMC8005094 DOI: 10.3390/molecules26061792] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 03/16/2021] [Accepted: 03/17/2021] [Indexed: 01/01/2023] Open
Abstract
Multimeric ligands consisting of multiple pharmacophores connected to a single backbone have been widely investigated for diagnostic and therapeutic applications. In this review, we summarize recent developments regarding multimeric radioligands targeting integrin αvβ3 receptors on cancer cells for molecular imaging and diagnostic applications using positron emission tomography (PET). Integrin αvβ3 receptors are glycoproteins expressed on the cell surface, which have a significant role in tumor angiogenesis. They act as receptors for several extracellular matrix proteins exposing the tripeptide sequence arginine-glycine-aspartic (RGD). Cyclic RDG peptidic ligands c(RGD) have been developed for integrin αvβ3 tumor-targeting positron emission tomography (PET) diagnosis. Several c(RGD) pharmacophores, connected with the linker and conjugated to a chelator or precursor for radiolabeling with different PET radionuclides (18F, 64Cu, and 68Ga), have resulted in multimeric ligands superior to c(RGD) monomers. The binding avidity, pharmacodynamic, and PET imaging properties of these multimeric c(RGD) radioligands, in relation to their structural characteristics are analyzed and discussed. Furthermore, specific examples from preclinical studies and clinical investigations are included.
Collapse
Affiliation(s)
- Christos Liolios
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Ag. Paraskevi Attikis, 15310 Athens, Greece
- Laboratory of Medicinal Chemistry, Department of Pharmacy, Section of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis–Zografou, 15771 Athens, Greece;
- Correspondence: (C.L.); (P.B.)
| | - Christos Sachpekidis
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (C.S.); (A.D.-S.)
| | - Antonios Kolocouris
- Laboratory of Medicinal Chemistry, Department of Pharmacy, Section of Pharmaceutical Chemistry, National and Kapodistrian University of Athens, Panepistimioupolis–Zografou, 15771 Athens, Greece;
| | - Antonia Dimitrakopoulou-Strauss
- Clinical Cooperation Unit Nuclear Medicine, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany; (C.S.); (A.D.-S.)
| | - Penelope Bouziotis
- Radiochemical Studies Laboratory, Institute of Nuclear & Radiological Sciences & Technology, Energy & Safety, National Centre for Scientific Research “Demokritos”, Ag. Paraskevi Attikis, 15310 Athens, Greece
- Correspondence: (C.L.); (P.B.)
| |
Collapse
|
31
|
Fu H, Du B, Chen Z, Li Y. Radiolabeled Peptides for SPECT and PET Imaging in the Detection of Breast Cancer: Preclinical and Clinical Perspectives. Curr Med Chem 2021; 27:6987-7002. [PMID: 32003658 DOI: 10.2174/0929867327666200128110827] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2019] [Revised: 12/22/2019] [Accepted: 01/02/2020] [Indexed: 01/05/2023]
Abstract
Breast cancer is the most common cancer in women worldwide. Due to the heterogeneous nature of breast cancer, the optimal treatment and expected response for each patient may not necessarily be universal. Molecular imaging techniques could play an important role in the early detection and targeted therapy evaluation of breast cancer. This review focuses on the development of peptides labeled with SPECT and PET radionuclides for breast cancer imaging. We summarized the current status of radiolabeled peptides for different receptors in breast cancer. The characteristics of radionuclides and major techniques for peptide labeling are also briefly discussed.
Collapse
Affiliation(s)
- Hao Fu
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Bulin Du
- Department of Nuclear Medicine, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zijun Chen
- Medical College of Xiamen University, Xiamen University, Xiamen, China
| | - Yesen Li
- Department of Nuclear Medicine & Minnan PET Center, Xiamen Cancer Hospital, The First Affiliated Hospital of Xiamen University, Teaching Hospital of Fujian Medical University, Xiamen, China
| |
Collapse
|
32
|
Shao T, Chen Z, Belov V, Wang X, Rwema SH, Kumar V, Fu H, Deng X, Rong J, Yu Q, Lang L, Lin W, Josephson L, Samir AE, Chen X, Chung RT, Liang SH. [ 18F]-Alfatide PET imaging of integrin αvβ3 for the non-invasive quantification of liver fibrosis. J Hepatol 2020; 73:161-169. [PMID: 32145257 PMCID: PMC7363052 DOI: 10.1016/j.jhep.2020.02.018] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Revised: 01/21/2020] [Accepted: 02/14/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND & AIMS The vitronectin receptor integrin αvβ3 drives fibrogenic activation of hepatic stellate cells (HSCs). Molecular imaging targeting the integrin αvβ3 could provide a non-invasive method for evaluating the expression and the function of the integrin αvβ3 on activated HSCs (aHSCs) in the injured liver. In this study, we sought to compare differences in the uptake of [18F]-Alfatide between normal and injured liver to evaluate its utility for assessment of hepatic fibrogenesis. METHODS PET with [18F]-Alfatide, non-enhanced CT, histopathology, immunofluorescence staining, immunoblotting and gene analysis were performed to evaluate and quantify hepatic integrin αvβ3 levels and liver fibrosis progression in mouse models of fibrosis (carbon tetrachloride [CCl4] and bile duct ligation [BDL]). The liver AUC divided by the blood AUC over 30 min was used as an integrin αvβ3-PET index to quantify fibrosis progression. Ex vivo analysis of frozen liver tissue from patients with fibrosis and cirrhosis verified the animal findings. RESULTS Fibrotic mouse livers showed enhanced [18F]-Alfatide uptake and retention compared to control livers. The radiotracer was demonstrated to bind specifically with integrin αvβ3, which is mainly expressed on aHSCs. Autoradiography and histopathology confirmed the PET imaging results. Further, the mRNA and protein level of integrin αvβ3 and its signaling complex were higher in CCl4 and BDL models than controls. The results obtained from analyses on human fibrotic liver sections supported the animal findings. CONCLUSIONS Imaging hepatic integrin αvβ3 with PET and [18F]-Alfatide offers a potential non-invasive method for monitoring the progression of liver fibrosis. LAY SUMMARY Integrin αvβ3 expression on activated hepatic stellate cells (aHSCs) is associated with HSC proliferation during hepatic fibrogenesis. Herein, we show that a radioactive tracer, [18F]-Alfatide, binds to integrin αvβ3 with high affinity and specificity. [18F]-Alfatide could thus be used as a non-invasive imaging biomarker to track hepatic fibrosis progression.
Collapse
Affiliation(s)
- Tuo Shao
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA; Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, USA
| | - Zhen Chen
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Vasily Belov
- Massachusetts General Hospital, Shriners Hospitals for Children, Boston, USA
| | - Xiaohong Wang
- Center for Ultrasound Research & Translation, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Steve H Rwema
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, USA
| | - Viksit Kumar
- Center for Ultrasound Research & Translation, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Hualong Fu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Xiaoyun Deng
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Jian Rong
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Qingzhen Yu
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Lixin Lang
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, USA
| | - Wenyu Lin
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, USA
| | - Lee Josephson
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Anthony E Samir
- Center for Ultrasound Research & Translation, Department of Radiology, Massachusetts General Hospital, Boston, USA
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine, National Institute of Biomedical Imaging and Bioengineering, National Institutes of Health, Bethesda, USA.
| | - Raymond T Chung
- Liver Center and Gastrointestinal Division, Department of Medicine, Massachusetts General Hospital, Boston, USA.
| | - Steven H Liang
- Division of Nuclear Medicine and Molecular Imaging, Department of Radiology, Massachusetts General Hospital, Boston, USA.
| |
Collapse
|
33
|
Liu Z, Yu L, Cheng K, Feng Y, Qiu P, Gai Y, Zhou M. Optimization, automation and validation of the large-scale radiosynthesis of Al 18F tracers in a custom-made automatic platform for high yield. REACT CHEM ENG 2020. [DOI: 10.1039/d0re00144a] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
A custom-made automatic platform was designed and developed for large scale Al18F tracer synthesis with high yield.
Collapse
Affiliation(s)
- Zhiguo Liu
- Department of PET/CT Center
- Shandong Cancer Hospital and Institute
- Shandong First Medical University and Shandong Academy of Medical Sciences
- Jinan
- China
| | - Lun Yu
- Department of PET-CT Center
- Chenzhou No. 1 People's Hospital
- Chenzhou 423000
- China
| | - Kai Cheng
- Department of PET/CT Center
- Shandong Cancer Hospital and Institute
- Shandong First Medical University and Shandong Academy of Medical Sciences
- Jinan
- China
| | - Yabo Feng
- Department of PET-CT Center
- Chenzhou No. 1 People's Hospital
- Chenzhou 423000
- China
| | - Pengfei Qiu
- Breast Cancer Center
- Shandong Cancer Hospital and Institute
- Shandong First Medical University and Shandong Academy of Medical Sciences
- Jinan 250117
- China
| | - Yongkang Gai
- Department of Nuclear Medicine
- Union Hospital
- Tongji Medical College
- Huazhong University of Science and Technology
- Hubei Province Key Laboratory of Molecular Imaging
| | - Ming Zhou
- Department of Nuclear Medicine
- Xiangya Hospital
- Central South University
- Changsha 410008
- China
| |
Collapse
|
34
|
Recent Advances in Nuclear Imaging of Receptor Expression to Guide Targeted Therapies in Breast Cancer. Cancers (Basel) 2019; 11:cancers11101614. [PMID: 31652624 PMCID: PMC6826563 DOI: 10.3390/cancers11101614] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2019] [Accepted: 10/18/2019] [Indexed: 12/12/2022] Open
Abstract
Breast cancer remains the most frequent cancer in women with different patterns of disease progression and response to treatments. The identification of specific biomarkers for different breast cancer subtypes has allowed the development of novel targeting agents for imaging and therapy. To date, patient management depends on immunohistochemistry analysis of receptor status on bioptic samples. This approach is too invasive, and in some cases, not entirely representative of the disease. Nuclear imaging using receptor tracers may provide whole-body information and detect any changes of receptor expression during disease progression. Therefore, imaging is useful to guide clinicians to select the best treatments for each patient and to evaluate early response thus reducing unnecessary therapies. In this review, we focused on the development of novel tracers that are ongoing in preclinical and/or clinical studies as promising tools to lead treatment decisions for breast cancer management.
Collapse
|
35
|
Fersing C, Bouhlel A, Cantelli C, Garrigue P, Lisowski V, Guillet B. A Comprehensive Review of Non-Covalent Radiofluorination Approaches Using Aluminum [ 18F]fluoride: Will [ 18F]AlF Replace 68Ga for Metal Chelate Labeling? Molecules 2019; 24:E2866. [PMID: 31394799 PMCID: PMC6719958 DOI: 10.3390/molecules24162866] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 07/31/2019] [Accepted: 08/06/2019] [Indexed: 12/20/2022] Open
Abstract
Due to its ideal physical properties, fluorine-18 turns out to be a key radionuclide for positron emission tomography (PET) imaging, for both preclinical and clinical applications. However, usual biomolecules radiofluorination procedures require the formation of covalent bonds with fluorinated prosthetic groups. This drawback makes radiofluorination impractical for routine radiolabeling, gallium-68 appearing to be much more convenient for the labeling of chelator-bearing PET probes. In response to this limitation, a recent expansion of the 18F chemical toolbox gave aluminum [18F]fluoride chemistry a real prominence since the late 2000s. This approach is based on the formation of an [18F][AlF]2+ cation, complexed with a 9-membered cyclic chelator such as NOTA, NODA or their analogs. Allowing a one-step radiofluorination in an aqueous medium, this technique combines fluorine-18 and non-covalent radiolabeling with the advantage of being very easy to implement. Since its first reports, [18F]AlF radiolabeling approach has been applied to a wide variety of potential PET imaging vectors, whether of peptidic, proteic, or small molecule structure. Most of these [18F]AlF-labeled tracers showed promising preclinical results and have reached the clinical evaluation stage for some of them. The aim of this report is to provide a comprehensive overview of [18F]AlF labeling applications through a description of the various [18F]AlF-labeled conjugates, from their radiosynthesis to their evaluation as PET imaging agents.
Collapse
Affiliation(s)
- Cyril Fersing
- Institut de Recherche en Cancérologie de Montpellier (IRCM), University of Montpellier, INSERM U1194, Montpellier Cancer Institute (ICM), 34298 Montpellier, France.
- Nuclear Medicine Department, Montpellier Cancer Institute (ICM), University of Montpellier, 208 Avenue des Apothicaires, 34298 Montpellier CEDEX 5, France.
| | - Ahlem Bouhlel
- CERIMED, Aix-Marseille University, 13005 Marseille, France
- Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM 1263, INRA 1260, 13385 Marseille, France
| | - Christophe Cantelli
- Institut de Recherche en Cancérologie de Montpellier (IRCM), University of Montpellier, INSERM U1194, Montpellier Cancer Institute (ICM), 34298 Montpellier, France
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, Université de Montpellier, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 34093 Montpellier CEDEX, France
| | - Philippe Garrigue
- CERIMED, Aix-Marseille University, 13005 Marseille, France
- Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM 1263, INRA 1260, 13385 Marseille, France
- Department of Nuclear Medicine, Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille (AP-HM), 13385 Marseille, France
| | - Vincent Lisowski
- Institut des Biomolécules Max Mousseron, UMR 5247, CNRS, Université de Montpellier, ENSCM, UFR des Sciences Pharmaceutiques et Biologiques, 34093 Montpellier CEDEX, France
| | - Benjamin Guillet
- CERIMED, Aix-Marseille University, 13005 Marseille, France
- Centre de recherche en CardioVasculaire et Nutrition (C2VN), Aix-Marseille University, INSERM 1263, INRA 1260, 13385 Marseille, France
- Department of Nuclear Medicine, Aix-Marseille University, Assistance Publique-Hôpitaux de Marseille (AP-HM), 13385 Marseille, France
| |
Collapse
|
36
|
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
|