1
|
Tesse G, Tolomeo A, De Filippis B, Giampietro L. Radiolabeled Probes from Derivatives of Natural Compounds Used in Nuclear Medicine. Molecules 2024; 29:4260. [PMID: 39275108 PMCID: PMC11396893 DOI: 10.3390/molecules29174260] [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: 07/24/2024] [Revised: 08/27/2024] [Accepted: 09/06/2024] [Indexed: 09/16/2024] Open
Abstract
Natural compounds are important precursors for the synthesis of new drugs. The development of novel molecules that are useful for various diseases is the main goal of researchers, especially for the diagnosis and treatment of many diseases. Some pathologies need to be treated with radiopharmaceuticals, and, for this reason, radiopharmaceuticals that use the radiolabeling of natural derivates molecules are arousing more and more interest. Radiopharmaceuticals can be used for both diagnostic and therapeutic purposes depending on the radionuclide. β+- and gamma-emitting radionuclides are used for diagnostic use for PET or SPECT imaging techniques, while α- and β--emitting radionuclides are used for in metabolic radiotherapy. Based on these assumptions, the purpose of this review is to highlight the studies carried out in the last ten years, to search for potentially useful radiopharmaceuticals for nuclear medicine that use molecules of natural origin as lead structures. In this context, the main radiolabeled compounds containing natural products as scaffolds are analyzed, in particular curcumin, stilbene, chalcone, and benzofuran. Studies on structural and chemical modifications are emphasized in order to obtain a collection of potential radiopharmaceuticals that exploit the biological properties of molecules of natural origin. The radionuclides used to label these compounds are 68Ga, 44Sc, 18F, 64Cu, 99mTc, and 125I for diagnostic imaging.
Collapse
Affiliation(s)
- Giuseppe Tesse
- Radiopharma Division, ITEL Telecomunicazioni s.r.l., 70037 Ruvo di Puglia, BA, Italy
| | - Anna Tolomeo
- Radiopharma Division, ITEL Telecomunicazioni s.r.l., 70037 Ruvo di Puglia, BA, Italy
| | - Barbara De Filippis
- Department of Pharmacy, Università degli Studi G. d'Annunzio, 66100 Chieti, CH, Italy
| | - Letizia Giampietro
- Department of Pharmacy, Università degli Studi G. d'Annunzio, 66100 Chieti, CH, Italy
| |
Collapse
|
2
|
Luke JJ, Davar D, Andtbacka RH, Bhardwaj N, Brody JD, Chesney J, Coffin R, de Baere T, de Gruijl TD, Fury M, Goldmacher G, Harrington KJ, Kaufman H, Kelly CM, Khilnani AD, Liu K, Loi S, Long GV, Melero I, Middleton M, Neyns B, Pinato DJ, Sheth RA, Solomon SB, Szapary P, Marabelle A. Society for Immunotherapy of Cancer (SITC) recommendations on intratumoral immunotherapy clinical trials (IICT): from premalignant to metastatic disease. J Immunother Cancer 2024; 12:e008378. [PMID: 38641350 PMCID: PMC11029323 DOI: 10.1136/jitc-2023-008378] [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] [Accepted: 02/22/2024] [Indexed: 04/21/2024] Open
Abstract
BACKGROUND Intratumorally delivered immunotherapies have the potential to favorably alter the local tumor microenvironment and may stimulate systemic host immunity, offering an alternative or adjunct to other local and systemic treatments. Despite their potential, these therapies have had limited success in late-phase trials for advanced cancer resulting in few formal approvals. The Society for Immunotherapy of Cancer (SITC) convened a panel of experts to determine how to design clinical trials with the greatest chance of demonstrating the benefits of intratumoral immunotherapy for patients with cancers across all stages of pathogenesis. METHODS An Intratumoral Immunotherapy Clinical Trials Expert Panel composed of international key stakeholders from academia and industry was assembled. A multiple choice/free response survey was distributed to the panel, and the results of this survey were discussed during a half-day consensus meeting. Key discussion points are summarized in the following manuscript. RESULTS The panel determined unique clinical trial designs tailored to different stages of cancer development-from premalignant to unresectable/metastatic-that can maximize the chance of capturing the effect of intratumoral immunotherapies. Design elements discussed included study type, patient stratification and exclusion criteria, indications of randomization, study arm determination, endpoints, biological sample collection, and response assessment with biomarkers and imaging. Populations to prioritize for the study of intratumoral immunotherapy, including stage, type of cancer and line of treatment, were also discussed along with common barriers to the development of these local treatments. CONCLUSIONS The SITC Intratumoral Immunotherapy Clinical Trials Expert Panel has identified key considerations for the design and implementation of studies that have the greatest potential to capture the effect of intratumorally delivered immunotherapies. With more effective and standardized trial designs, the potential of intratumoral immunotherapy can be realized and lead to regulatory approvals that will extend the benefit of these local treatments to the patients who need them the most.
Collapse
Affiliation(s)
- Jason J Luke
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | - Diwakar Davar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Nina Bhardwaj
- Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Joshua D Brody
- Marc and Jennifer Lipschultz Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, USA
| | - Jason Chesney
- James Graham Brown Cancer Center, University of Louisville, Louisville, Kentucky, USA
| | | | - Thierry de Baere
- Center for Biotherapies In Situ (BIOTHERIS), INSERM CIC1428, Interventional Radiology Unit, Department of Medical Imaging, Gustave Roussy Cancer Center, University of Paris Saclay, Villejuif, France
| | - Tanja D de Gruijl
- Department of Medical Oncology, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, Netherlands
- Cancer Biology and Immunology, Cancer Center Amsterdam, Amsterdam, Netherlands
- Cancer Immunology, Amsterdam Institute for Infection and Immunology, Amsterdam, Netherlands
| | - Matthew Fury
- Oncology Clinical Development, Regeneron Pharmaceuticals Inc, Tarrytown, New York, USA
| | | | - Kevin J Harrington
- The Institute of Cancer Research, The Royal Marsden National Institute for Health and Care Research Biomedical Research Centre, London, UK
| | - Howard Kaufman
- Department of Surgery, Massachusetts General Hospital, Boston, Massachusetts, USA
- Ankyra Therapeutics, Boston, Massachusetts, USA
| | - Ciara M Kelly
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | | | - Ke Liu
- Marengo Therapeutics, Inc, Cambridge, Massachusetts, USA
| | - Sherene Loi
- Division of Cancer Research, Peter MacCallum Cancer Center, Melbourne, Victoria, Australia
- The Sir Peter MacCallum Department of Oncology, The University of Melbourne, Melbourne, Victoria, Australia
| | - Georgina V Long
- Melanoma Institute Australia, University of Sydney, and Royal North Shore and Mater Hospitals, North Sydney, New South Wales, Australia
| | | | - Mark Middleton
- Department of Oncology, University of Oxford, Oxford, UK
| | - Bart Neyns
- Department of Medical Oncology, Universitair Ziekenhuis Brussel, Jette, Belgium
| | - David J Pinato
- Department of Surgery and Cancer, Imperial College London, Hammersmith Hospital, London, UK
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| | - Rahul A Sheth
- Department of Interventional Radiology, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen B Solomon
- Chief of Interventional Radiology, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Professor of Radiology, Weill Cornell Medical College, New York, New York, USA
| | - Philippe Szapary
- Interventional Oncology, Johnson & Johnson, New Brunswick, New Jersey, USA
| | - Aurelien Marabelle
- Center for Biotherapies In Situ (BIOTHERIS), INSERM CIC1428, Department for Therapeutic Innovation and Early Phase Trials (DITEP), Gustave Roussy Cancer Center, University of Paris Saclay, Villejuif, France
| |
Collapse
|
3
|
Garcia N, Ulin M, Yang Q, Ali M, Bosland MC, Zeng W, Chen L, Al-Hendy A. Survivin-Sodium Iodide Symporter Reporter as a Non-Invasive Diagnostic Marker to Differentiate Uterine Leiomyosarcoma from Leiomyoma. Cells 2023; 12:2830. [PMID: 38132150 PMCID: PMC10741838 DOI: 10.3390/cells12242830] [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: 10/23/2023] [Revised: 12/04/2023] [Accepted: 12/08/2023] [Indexed: 12/23/2023] Open
Abstract
Leiomyosarcoma (LMS) has been challenging to diagnose because of limitations in clinical and radiographic predictors, as well as the lack of reliable serum or urinary biomarkers. Most uterine masses consist of benign leiomyoma (LM). However, it is currently a significant challenge in gynecology practice to differentiate LMS from LM. This inability poses grave consequences for patients, leading to a high number of unnecessary hysterectomies, infertility, and other major morbidities and possible mortalities. This study aimed to evaluate the use of Survivin-Sodium iodide symporter (Ad-Sur-NIS) as a reporter gene biomarker to differentiate malignant LMS from benign LM by using an F18-NaBF4 PET/CT scan. The PET/CT scan images showed a significantly increased radiotracer uptake and a decreased radiotracer decay attributable to the higher abundance of Ad-Sur-NIS in the LMS tumors compared to LM (p < 0.05). An excellent safety profile was observed, with no pathological or metabolic differences detected in Ad-Sur-NIS-treated animal versus the vehicle control. Ad-Sur-NIS as a PET scan reporter is a promising imaging biomarker that can differentiate uterine LMS from LM using F18-NaBF4 as a radiotracer. As a new diagnostic method, the F18 NaBF4 PET/CT scan can provide a much-needed tool in clinical practices to effectively triage women with suspicious uterine masses and avoid unnecessary invasive interventions.
Collapse
Affiliation(s)
- Natalia Garcia
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Greehey Children’s Cancer Research Institute, The University of Texas Health Science Center, San Antonio, TX 77030, USA
| | - Mara Ulin
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, Mount Sinai Hospital, Chicago, IL 11537, USA
| | - Qiwei Yang
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| | - Mohamed Ali
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
- Clinical Pharmacy Department, Faculty of Pharmacy, Ain Shams University, Cairo 11566, Egypt
| | - Maarten C. Bosland
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60607, USA;
| | - Weiqiao Zeng
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
| | - Liaohai Chen
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
| | - Ayman Al-Hendy
- Department of Surgery, University of Illinois at Chicago, Chicago, IL 60607, USA; (N.G.); (M.U.); (Q.Y.); (M.A.); (W.Z.); (L.C.)
- Department of Obstetrics and Gynecology, University of Chicago, Chicago, IL 60637, USA
| |
Collapse
|
4
|
Akbari B, Huber BR, Sherman JH. Unlocking the Hidden Depths: Multi-Modal Integration of Imaging Mass Spectrometry-Based and Molecular Imaging Techniques. Crit Rev Anal Chem 2023; 55:109-138. [PMID: 37847593 DOI: 10.1080/10408347.2023.2266838] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2023]
Abstract
Multimodal imaging (MMI) has emerged as a powerful tool in clinical research, combining different imaging modes to acquire comprehensive information and enabling scientists and surgeons to study tissue identification, localization, metabolic activity, and molecular discovery, thus aiding in disease progression analysis. While multimodal instruments are gaining popularity, challenges such as non-standardized characteristics, custom software, inadequate commercial support, and integration issues with other instruments need to be addressed. The field of multimodal imaging or multiplexed imaging allows for simultaneous signal reproduction from multiple imaging strategies. Intraoperatively, MMI can be integrated into frameless stereotactic surgery. Recent developments in medical imaging modalities such as magnetic resonance imaging (MRI), and Positron Emission Topography (PET) have brought new perspectives to multimodal imaging, enabling early cancer detection, molecular tracking, and real-time progression monitoring. Despite the evidence supporting the role of MMI in surgical decision-making, there is a need for comprehensive studies to validate and perform integration at the intersection of multiple imaging technologies. They were integrating mass spectrometry-based technologies (e.g., imaging mass spectrometry (IMS), imaging mass cytometry (IMC), and Ion mobility mass spectrometry ((IM-IM) with medical imaging modalities, offering promising avenues for molecular discovery and clinical applications. This review emphasizes the potential of multi-omics approaches in tissue mapping using MMI integrated into desorption electrospray ionization (DESI) and matrix-assisted laser desorption ionization (MALDI), allowing for sequential analyses of the same section. By addressing existing knowledge gaps, this review encourages future research endeavors toward multi-omics approaches, providing a roadmap for future research and enhancing the value of MMI in molecular pathology for diagnosis.
Collapse
Affiliation(s)
- Behnaz Akbari
- Department of Chemistry, Purdue University, West Lafayette, Indiana, USA
| | - Bertrand Russell Huber
- Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
- Boston University Alzheimer's Disease and CTE Center, Boston University School of Medicine, Boston, Massachusetts, USA
- Department of Neurology, Boston University School of Medicine, Boston, Massachusetts, USA
- US Department of Veteran Affairs, VA Boston Healthcare System, Boston, Massachusetts USA
- US Department of Veterans Affairs, National Center for PTSD, Boston, Massachusetts USA
| | - Janet Hope Sherman
- Chobanian and Avedisian School of Medicine, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
5
|
Sullivan TE, Hernandez Vargas S, Ghosh SC, AghaAmiri S, Ikoma N, Azhdarinia A. A translational blueprint for developing intraoperative imaging agents via radiopharmaceutical-guided drug design. Curr Opin Chem Biol 2023; 76:102376. [PMID: 37572489 DOI: 10.1016/j.cbpa.2023.102376] [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: 02/09/2023] [Revised: 06/22/2023] [Accepted: 07/07/2023] [Indexed: 08/14/2023]
Abstract
Cancer imaging is a rapidly evolving field due to the discovery of novel molecular targets and the availability of corresponding techniques to detect them with high precision, accuracy, and sensitivity. Nuclear medicine is the most widely used molecular imaging modality and has a growing toolkit of clinically used radiopharmaceuticals that enable whole-body tumor visualization, staging, and treatment monitoring for a variety of tumors in a non-invasive manner. The need for similar imaging capabilities in the operating room has led to the emergence of fluorescence-guided surgery (FGS) as a powerful technique that gives surgeons unprecedented ability to distinguish tumors from healthy tissues. While a variety of strategies have been used to develop contrast agents for FGS, the use of radiopharmaceuticals as models brings exceptional translational potential and has increasingly been explored. Here, we review strategies used to convert clinically used radiopharmaceuticals into fluorescent and multimodal counterparts. Unique preclinical and clinical capabilities stemming from radiopharmaceutical-based agent design are also discussed to illustrate the advantages of this approach.
Collapse
Affiliation(s)
- Teresa E Sullivan
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Servando Hernandez Vargas
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Sukhen C Ghosh
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Solmaz AghaAmiri
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA
| | - Naruhiko Ikoma
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Houston, TX, 77030, USA
| | - Ali Azhdarinia
- The Brown Foundation Institute of Molecular Medicine, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX 77054, USA.
| |
Collapse
|
6
|
Xu Y, Cen P, Ma L, Tian M, Zhang X, Zhang Q, Yu K, Zhang H, Gu W, He Q. Highly efficient radiosynthesis and biological evaluation of [18F]safinamide, a radiolabelled anti-parkinsonian drug for PET imaging. ChemMedChem 2022; 17:e202200472. [PMID: 36068922 DOI: 10.1002/cmdc.202200472] [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: 08/31/2022] [Indexed: 11/06/2022]
Abstract
As an add-on drug approved for Parkinson's disease treatment, safinamide has multiple functions, such as selective and reversible monoamine oxidase-B inhibition, voltage-sensitive sodium/potassium channel blockage, and glutamate release inhibition. Meanwhile, safinamide shows tremendous therapeutic potential in the context of other central nervous system diseases (e.g., ischaemic stroke, amyotrophic lateral sclerosis, depression, etc.). In this work, [18F]safinamide, which is safinamide labelled by the positron-emitting radionuclide [18F]fluorine, was synthesized automatically based on iodonium ylide precursors with high radiochemical yield and high molar activity. Density functional theory was applied to calculate the Gibbs free energy change during iodonium ylide-mediated fluorination and to interpret the effect of tetraethylammonium (TEA+) as the counter cation in these reactions to improve the nucleophilicity of [18F/19F]fluoride. In addition, positron emission tomography studies on Sprague Dawley rats were carried out to determine the imaging characteristics, pharmacokinetics, and metabolism of the [18F]safinamide radiotracer. The results displayed the complete biodistribution of the radiotracer, especially in rat brains, and revealed that [18F]safinamide has moderate brain uptake, rapid and reversible binding kinetics, and good stability.
Collapse
Affiliation(s)
- Yangyang Xu
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Peili Cen
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Lijuan Ma
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Mei Tian
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Xue Zhang
- Chinese Academy of Sciences, Shenzhen Institutes of Advanced Technology, CHINA
| | - Qinghua Zhang
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Kaiwu Yu
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Hong Zhang
- Zhejiang University, Department of Nuclear Medicine and PET/CT Center, CHINA
| | - Wangjun Gu
- Zhejiang University, College of Chemical & Biological Engineering, CHINA
| | - Qinggang He
- Zhejiang University, Chemical Engineering, 38 Zheda Rd., 310027, Hangzhou, CHINA
| |
Collapse
|
7
|
New Imaging Modality of COVID-19 Pneumonia Developed on the Basis of Alzheimer’s Disease Research. Int J Mol Sci 2022; 23:ijms23158405. [PMID: 35955536 PMCID: PMC9369300 DOI: 10.3390/ijms23158405] [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: 06/14/2022] [Revised: 07/20/2022] [Accepted: 07/27/2022] [Indexed: 11/30/2022] Open
Abstract
Viral pneumonia caused by highly infectious SARS-CoV-2 poses a higher risk to older people and those who have underlying health conditions, including Alzheimer’s disease. In this work we present newly designed tacrine-based radioconjugates with physicochemical and biological properties that are crucial for the potential application as diagnostic radiopharmaceuticals. A set of ten tacrine derivatives was synthesized, labelled with gallium-68 and fully characterized in the context of their physicochemical properties. Based on these results, the final two most promising radioconjugates, [68Ga]Ga-NODAGA-Bn-NH(CH2)9Tac and [68Ga]Ga-THP-NH(CH2)9Tac, were selected for biodistribution studies. The latter compound was proven to be a good inhibitor of cholinesterases with significant affinity toward the lungs, according to the biodistribution studies. On the basis of molecular modelling combined with in vitro studies, we unraveled which structural properties of the developed tacrine derivatives are crucial for high affinity toward acetylcholinesterase, whose increased levels in lung tissues in the course of coronavirus disease indicate the onset of pneumonia. The radiopharmaceutical [68Ga]Ga-THP-NH(CH2)9Tac was ultimately selected due to its increased accuracy and improved sensitivity in PET imaging of lung tissue with high levels of acetylcholinesterase, and it may become a novel potential diagnostic modality for the determination of lung perfusion, including in inflammation after COVID-19.
Collapse
|
8
|
Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis. Int J Mol Sci 2022; 23:ijms23137071. [PMID: 35806074 PMCID: PMC9267012 DOI: 10.3390/ijms23137071] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2022] [Revised: 06/23/2022] [Accepted: 06/23/2022] [Indexed: 12/14/2022] Open
Abstract
Extensive angiogenesis is a characteristic feature in the synovial tissue of rheumatoid arthritis (RA) from a very early stage of the disease onward and constitutes a crucial event for the development of the proliferative synovium. This process is markedly intensified in patients with prolonged disease duration, high disease activity, disease severity, and significant inflammatory cell infiltration. Angiogenesis is therefore an interesting target for the development of new therapeutic approaches as well as disease monitoring strategies in RA. To this end, nuclear imaging modalities represent valuable non-invasive tools that can selectively target molecular markers of angiogenesis and accurately and quantitatively track molecular changes in multiple joints simultaneously. This systematic review summarizes the imaging markers used for single photon emission computed tomography (SPECT) and/or positron emission tomography (PET) approaches, targeting pathways and mediators involved in synovial neo-angiogenesis in RA.
Collapse
|
9
|
Burt T, Roffel AF, Langer O, Anderson K, DiMasi J. Strategic, feasibility, economic, and cultural aspects of phase 0 approaches: Is it time to change the drug development process in order to increase productivity? Clin Transl Sci 2022; 15:1355-1379. [PMID: 35278281 PMCID: PMC9199889 DOI: 10.1111/cts.13269] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2021] [Revised: 01/20/2022] [Accepted: 02/28/2022] [Indexed: 12/05/2022] Open
Abstract
Research conducted over the past 2 decades has enhanced the validity and expanded the applications of microdosing and other phase 0 approaches in drug development. Phase 0 approaches can accelerate drug development timelines and reduce attrition in clinical development by increasing the quality of candidates entering clinical development and by reducing the time to "go-no-go" decisions. This can be done by adding clinical trial data (both healthy volunteers and patients) to preclinical candidate selection, and by applying methodological and operational advantages that phase 0 have over traditional approaches. The main feature of phase 0 approaches is the limited, subtherapeutic exposure to the test article. This means a reduced risk to research volunteers, and reduced regulatory requirements, timelines, and costs of first-in-human (FIH) testing. Whereas many operational aspects of phase 0 approaches are similar to those of other early phase clinical development programs, they have some unique strategic, regulatory, ethical, feasibility, economic, and cultural aspects. Here, we provide a guidance to these operational aspects and include case studies to highlight their potential impact in a range of clinical development scenarios.
Collapse
Affiliation(s)
- Tal Burt
- Phase-0/Microdosing Network, New York, New York, USA
- Burt Consultancy, LLC, New York, New York, USA
| | | | - Oliver Langer
- Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria
- Department of Biomedical Imaging and Image-guided Therapy, Medical University of Vienna, Vienna, Austria
| | | | - Joseph DiMasi
- Tufts Center for the Study of Drug Development, Tufts University, Boston, Massachusetts, USA
| |
Collapse
|
10
|
Jaswal A, Hazari PP, Prakash S, Sethi P, Kaushik A, Roy BG, Kathait S, Singh B, Mishra AK. [ 99mTc]Tc-DTPA-Bis(cholineethylamine) as an Oncologic Tracer for the Detection of Choline Transporter (ChT) and Choline Kinase (ChK) Expression in Cancer. ACS OMEGA 2022; 7:12509-12523. [PMID: 35474820 PMCID: PMC9025991 DOI: 10.1021/acsomega.1c04256] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Accepted: 03/08/2022] [Indexed: 06/14/2023]
Abstract
OBJECTIVE The elevated choline transporters (ChT), choline kinase (ChK), choline uptake, and phosphorylation in certain tumor cells have influenced the development of radiolabeled choline derivatives as diagnostic probes for imaging cell membrane proliferation. We, therefore, aimed to develop a choline-based moiety for imaging choline kinase-overexpressed tumors by single-photon emission tomography (SPECT). A novel choline-based diagnostic probe was synthesized and evaluated preclinically in various ChT- and ChK-overexpressed tumor models for SPECT imaging applications. METHODS The synthesis of diethylenetriaminepentaacetic acid-bis-choline ethylamine [DTPA-bis(ChoEA)] featured the conjugation of dimethylaminoethanol to a bifunctional chelator DTPA anhydride. [99mTc]Tc-DTPA-bis(ChoEA) was prepared, and its in vivo characteristics were evaluated in BALB/c mice and tumor-xenografted PC3, A549, and HCT116 athymic mouse models. The in vitro parameters, including cell binding and cytotoxicity, were assessed in PC3, A549, and HCT116 cell lines. To evaluate the specificity of the radioprobe, competitive binding studies were performed. Small-animal SPECT/CT diagnostic imaging was performed for in vivo evaluation. The mouse biodistribution data was further investigated to estimate the radiation dose in humans. RESULTS In silico studies suggested high binding with enhanced specificity. A standard radiolabeling procedure using stannous chloride as a reducing agent showed a labeling yield of 99.5 ± 0.5%. The in silico studies suggested high binding with enhanced specificity. [99mTc]Tc-DTPA-bis(ChoEA) showed high in vitro stability and specificity. The pharmacokinetic studies of [99mTc]Tc-DTPA-bis(ChoEA) in mice showed an increased tumor-to-background ratio after few minutes of intravenous administration. The first-in-human trial was also conducted. The effective dose was estimated to be 0.00467 mSv/MBq (4.67 mSv/GBq), resulting in a radiation dose of up to 1.73 mSv for the 370 MBq injection of [99mTc]Tc-DTPA-bis(ChoEA). CONCLUSIONS The synthesized radioprobe [99mTc]Tc-DTPA-bis(ChoEA) accumulates specifically in choline kinase-overexpressed tumors with a high signal-to-noise ratio. The preclinical and first-in-man data suggested that [99mTc]Tc-DTPA-bis(ChoEA) could potentially be used as a diagnostic SPECT tracer in the monitoring and staging of cancer.
Collapse
Affiliation(s)
- Ambika
Parmar Jaswal
- Division
of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Puja Panwar Hazari
- Division
of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Surbhi Prakash
- Division
of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Pallavi Sethi
- Division
of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Aruna Kaushik
- Department
of Nuclear Medicine, Institute of Nuclear
Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Bal G. Roy
- Experimental
Animal Facility, Institute of Nuclear Medicine
and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Swati Kathait
- Division
of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| | - Baljinder Singh
- Post
Graduate Institute of Medical Education & Research, Chandigarh 160012, India
| | - Anil Kumar Mishra
- Division
of Cyclotron and Radiopharmaceutical Sciences, Institute of Nuclear Medicine and Allied Sciences, Brig S.K. Mazumdar Road, Delhi 110054, India
| |
Collapse
|
11
|
Pisaneschi F, Viola NT. Development and Validation of a PET/SPECT Radiopharmaceutical in Oncology. Mol Imaging Biol 2022; 24:1-7. [PMID: 34542804 PMCID: PMC8760224 DOI: 10.1007/s11307-021-01645-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Revised: 08/17/2021] [Accepted: 08/18/2021] [Indexed: 01/10/2023]
Abstract
In oncology, biomarker research aimed to provide insights on cancer biology via positron emission tomography (PET) and single photon emission tomography (SPECT) imaging has seen an incredible growth in the past two decades. Despite the increased number of publications on PET/SPECT radiopharmaceuticals, the field lacked standardization of in vitro and in vivo parameters necessary for the characterization of any radiotracer. Through the efforts of the World Molecular Imaging Society Education Committee, this white paper lays down validation studies that are essential to chemically and biologically characterize new radiopharmaceuticals derived from small molecules, peptides or proteins. Finally, a brief overview of the steps toward translation is also presented.Herein, we discuss the following: Chemistry and radiochemistry metrics to establish the identity of the imaging agent. In vitro and in vivo studies to examine the radiotracer's mechanism of action, which includes target specificity, pharmacokinetics and in vivo metabolism.
Collapse
Affiliation(s)
- Federica Pisaneschi
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, TX 77030 USA
| | - Nerissa T. Viola
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201 USA
| |
Collapse
|
12
|
McAteer MA, O'Connor JPB, Koh DM, Leung HY, Doran SJ, Jauregui-Osoro M, Muirhead N, Brew-Graves C, Plummer ER, Sala E, Ng T, Aboagye EO, Higgins GS, Punwani S. Introduction to the National Cancer Imaging Translational Accelerator (NCITA): a UK-wide infrastructure for multicentre clinical translation of cancer imaging biomarkers. Br J Cancer 2021; 125:1462-1465. [PMID: 34316019 PMCID: PMC8313668 DOI: 10.1038/s41416-021-01497-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Accepted: 07/09/2021] [Indexed: 12/14/2022] Open
Abstract
The National Cancer Imaging Translational Accelerator (NCITA) is creating a UK national coordinated infrastructure for accelerated translation of imaging biomarkers for clinical use. Through the development of standardised protocols, data integration tools and ongoing training programmes, NCITA provides a unique scalable infrastructure for imaging biomarker qualification using multicentre clinical studies.
Collapse
Affiliation(s)
- M A McAteer
- Department of Oncology, University of Oxford, Oxford, UK.
| | - J P B O'Connor
- Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - D M Koh
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - H Y Leung
- Beatson Institute for Cancer Research, University of Glasgow, Glasgow, UK
| | - S J Doran
- Division of Radiotherapy and Imaging, The Institute of Cancer Research and Royal Marsden Hospital, London, UK
| | - M Jauregui-Osoro
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - N Muirhead
- Centre for Medical Imaging, University College London, London, UK
| | - C Brew-Graves
- Centre for Medical Imaging, University College London, London, UK
| | - E R Plummer
- Northern Institute for Cancer Care, Freeman Hospital and Newcastle University, Newcastle upon Tyne, UK
| | - E Sala
- Department of Radiology, University of Cambridge and CRUK Cambridge Centre, Cambridge, UK
| | - T Ng
- UCL Cancer Institute, University College London, London, UK
- School of Cancer and Pharmaceutical Sciences, King's College London, London, UK
| | - E O Aboagye
- Department of Surgery and Cancer, Imperial College London, London, UK
| | - G S Higgins
- Department of Oncology, University of Oxford, Oxford, UK
| | - S Punwani
- Centre for Medical Imaging, University College London, London, UK
| |
Collapse
|
13
|
Britton R, Gouverneur V, Lin JH, Meanwell M, Ni C, Pupo G, Xiao JC, Hu J. Contemporary synthetic strategies in organofluorine chemistry. ACTA ACUST UNITED AC 2021. [DOI: 10.1038/s43586-021-00042-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
14
|
Wongso H. Natural product-based Radiopharmaceuticals:Focus on curcumin and its analogs, flavonoids, and marine peptides. J Pharm Anal 2021; 12:380-393. [PMID: 35811617 PMCID: PMC9257450 DOI: 10.1016/j.jpha.2021.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 05/19/2021] [Accepted: 07/19/2021] [Indexed: 02/07/2023] Open
Abstract
Natural products provide a bountiful supply of pharmacologically relevant precursors for the development of various drug-related molecules, including radiopharmaceuticals. However, current knowledge regarding the importance of natural products in developing new radiopharmaceuticals remains limited. To date, several radionuclides, including gallium-68, technetium-99m, fluorine-18, iodine-131, and iodine-125, have been extensively studied for the synthesis of diagnostic and therapeutic radiopharmaceuticals. The availability of various radiolabeling methods allows the incorporation of these radionuclides into bioactive molecules in a practical and efficient manner. Of the radiolabeling methods, direct radioiodination, radiometal complexation, and halogenation are generally suitable for natural products owing to their simplicity and robustness. This review highlights the pharmacological benefits of curcumin and its analogs, flavonoids, and marine peptides in treating human pathologies and provides a perspective on the potential use of these bioactive compounds as molecular templates for the design and development of new radiopharmaceuticals. Additionally, this review provides insights into the current strategies for labeling natural products with various radionuclides using either direct or indirect methods. Potential use of natural products for the development of diagnostic and therapeutic radiopharmaceuticals. Profile of potential natural products as molecular templates for the synthesis of new radiopharmaceuticals: Focus on curcumin and its closely related substances, flavonoids, and marine peptides. Radiolabeling strategies, challenges, and examples of natural product-based radiopharmaceuticals under investigation.
Collapse
|
15
|
Langer LB, Hess A, Korkmaz Z, Tillmanns J, Reffert LM, Bankstahl JP, Bengel FM, Thackeray JT, Ross TL. Molecular imaging of fibroblast activation protein after myocardial infarction using the novel radiotracer [ 68Ga]MHLL1. Am J Cancer Res 2021; 11:7755-7766. [PMID: 34335962 PMCID: PMC8315078 DOI: 10.7150/thno.51419] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Accepted: 05/11/2021] [Indexed: 11/24/2022] Open
Abstract
Background: Myocardial infarction (MI) evokes an organized remodeling process characterized by the activation and transdifferentiation of quiescent cardiac fibroblasts to generate a stable collagen rich scar. Early fibroblast activation may be amenable to targeted therapy, but is challenging to identify in vivo. We aimed to non-invasively image active fibrosis by targeting the fibroblast activation protein (FAP) expressed by activated (myo)fibroblasts, using a novel positron emission tomography (PET) radioligand [68Ga]MHLL1 after acute MI. Methods: One-step chemical synthesis and manual as well as module-based radiolabeling yielded [68Ga]MHLL1. Binding characteristics were evaluated in murine and human FAP-transfected cells, and stability tested in human serum. Biodistribution in healthy animals was interrogated by dynamic PET imaging, and metabolites were measured in blood and urine. The temporal pattern of FAP expression was determined by serial PET imaging at 7 d and 21 d after coronary artery ligation in mice as percent injected dose per gram (%ID/g). PET measurements were validated by ex vivo autoradiography and immunostaining for FAP and inflammatory macrophages. Results: [68Ga]MHLL1 displayed specific uptake in murine and human FAP-positive cells (p = 0.0208). In healthy mice the tracer exhibited favorable imaging characteristics, with low blood pool retention and dominantly renal clearance. At 7 d after coronary artery ligation, [68Ga]MHLL1 uptake was elevated in the infarct relative to the non-infarcted remote myocardium (1.3 ± 0.3 vs. 1.0 ± 0.2 %ID/g, p < 0.001) which persisted to 21 d after MI (1.3 ± 0.4 vs. 1.1 ± 0.4 %ID/g, p = 0.013). Excess unlabeled compound blocked tracer accumulation in both infarct and non-infarct remote myocardium regions (p < 0.001). Autoradiography and histology confirmed the regional uptake of [68Ga]MHLL1 in the infarct and especially border zone regions, as identified by Masson trichrome collagen staining. Immunostaining further delineated persistent FAP expression at 7 d and 21 d post-MI in the border zone, consistent with tracer distribution in vivo. Conclusion: The simplified synthesis of [68Ga]MHLL1 bears promise for non-invasive characterization of fibroblast activation protein early in remodeling after MI.
Collapse
|
16
|
Liberini V, Laudicella R, Capozza M, Huellner MW, Burger IA, Baldari S, Terreno E, Deandreis D. The Future of Cancer Diagnosis, Treatment and Surveillance: A Systemic Review on Immunotherapy and Immuno-PET Radiotracers. Molecules 2021; 26:2201. [PMID: 33920423 PMCID: PMC8069316 DOI: 10.3390/molecules26082201] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/07/2021] [Accepted: 04/09/2021] [Indexed: 12/12/2022] Open
Abstract
Immunotherapy is an effective therapeutic option for several cancers. In the last years, the introduction of checkpoint inhibitors (ICIs) has shifted the therapeutic landscape in oncology and improved patient prognosis in a variety of neoplastic diseases. However, to date, the selection of the best patients eligible for these therapies, as well as the response assessment is still challenging. Patients are mainly stratified using an immunohistochemical analysis of the expression of antigens on biopsy specimens, such as PD-L1 and PD-1, on tumor cells, on peritumoral immune cells and/or in the tumor microenvironment (TME). Recently, the use and development of imaging biomarkers able to assess in-vivo cancer-related processes are becoming more important. Today, positron emission tomography (PET) with 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) is used routinely to evaluate tumor metabolism, and also to predict and monitor response to immunotherapy. Although highly sensitive, FDG-PET in general is rather unspecific. Novel radiopharmaceuticals (immuno-PET radiotracers), able to identify specific immune system targets, are under investigation in pre-clinical and clinical settings to better highlight all the mechanisms involved in immunotherapy. In this review, we will provide an overview of the main new immuno-PET radiotracers in development. We will also review the main players (immune cells, tumor cells and molecular targets) involved in immunotherapy. Furthermore, we report current applications and the evidence of using [18F]FDG PET in immunotherapy, including the use of artificial intelligence (AI).
Collapse
MESH Headings
- Antineoplastic Agents, Immunological/therapeutic use
- Artificial Intelligence
- B7-H1 Antigen/genetics
- B7-H1 Antigen/immunology
- Fluorodeoxyglucose F18/administration & dosage
- Fluorodeoxyglucose F18/chemistry
- Gene Expression Regulation, Neoplastic/drug effects
- Humans
- Immune Checkpoint Inhibitors/chemistry
- Immune Checkpoint Inhibitors/metabolism
- Immunotherapy, Adoptive/methods
- Killer Cells, Natural/drug effects
- Killer Cells, Natural/immunology
- Killer Cells, Natural/pathology
- Neoplasms/diagnostic imaging
- Neoplasms/genetics
- Neoplasms/immunology
- Neoplasms/therapy
- Positron-Emission Tomography/methods
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/immunology
- Radiopharmaceuticals/administration & dosage
- Radiopharmaceuticals/chemical synthesis
- Signal Transduction
- T-Lymphocytes, Cytotoxic/drug effects
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/pathology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/pathology
- Tumor Microenvironment/drug effects
- Tumor Microenvironment/genetics
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Virginia Liberini
- Department of Medical Science, Division of Nuclear Medicine, University of Torino, 10126 Torino, Italy;
| | - Riccardo Laudicella
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (S.B.)
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
| | - Martina Capozza
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Martin W. Huellner
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
| | - Irene A. Burger
- Department of Nuclear Medicine, University Hospital Zurich, University of Zurich, 8006 Zurich, Switzerland; (M.W.H.); (I.A.B.)
- Department of Nuclear Medicine, Kantonsspital Baden, 5004 Baden, Switzerland
| | - Sergio Baldari
- Department of Biomedical and Dental Sciences and of Morpho-Functional Imaging, Nuclear Medicine Unit, University of Messina, 98125 Messina, Italy; (R.L.); (S.B.)
| | - Enzo Terreno
- Molecular & Preclinical Imaging Centers, Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, 10126 Torino, Italy; (M.C.); (E.T.)
| | - Désirée Deandreis
- Department of Medical Science, Division of Nuclear Medicine, University of Torino, 10126 Torino, Italy;
| |
Collapse
|
17
|
Cabrera M, Lecot N, Fernández M, Gambini JP, Porcal W, Cabral P. 99mTc Stearyl 6-(benzylidenehydrazinyl) nicotinamide Liposomes as Tumor Permeability Evaluation Tracer. AAPS PharmSciTech 2021; 22:115. [PMID: 33763814 DOI: 10.1208/s12249-021-01984-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Accepted: 03/08/2021] [Indexed: 12/13/2022] Open
Abstract
Nanomedicine is a highly demanded discipline. Liposomes have seen an increased attention due to their physicochemical properties that allow them to act as nanocarriers of drugs and also of radioisotopes that can be used to diagnose and treat cancer. In order to obtain a novel permeability cancer imaging agent based on 99mTc-labeled liposomes, we describe microwave-assisted synthesis of stearyl 6-(benzylidenehydrazinyl) nicotinamide lipid, which was included in two formulations: nanometric hydrazinonicotinic acid (HYNIC) liposome and its PEGylated coated analogue, HYNIC-PEG liposome. Radiolabeling with 99mTc via stearyl 6-(benzylidenehydrazinyl) nicotinamide was found to be easy, reproducible, and stable, revealing high radiochemical purity (94 ± 1.7%) for both liposomal formulations. Biodistribution at 4 h and 24 h and scintigraphic images at 4 h were performed in normal and melanoma-bearing C57BL/6 mice. Biodistribution studies at 4 h showed tumor uptake of 99mTc-HYNIC liposome and 99mTc-HYNIC-PEG liposome (1.1 ± 0.6 and 2.5 ± 0.4, respectively) and also at 24 h p.i. (1.8 ± 0.5 and 3.0 ± 1.1, respectively). Scintigraphic images showed appreciable tumor uptake in melanoma tumor-bearing mice with both liposomal formulations. Our results show that 99mTc stearyl 6-(benzylidenehydrazinyl) nicotinamide liposomes can be used as diagnostic noninvasive in vivo tumor-targeting agents capable of evaluating tumor permeability and development who can be used in personalized chemotherapy planning.
Collapse
|
18
|
Meikle SR, Sossi V, Roncali E, Cherry SR, Banati R, Mankoff D, Jones T, James M, Sutcliffe J, Ouyang J, Petibon Y, Ma C, El Fakhri G, Surti S, Karp JS, Badawi RD, Yamaya T, Akamatsu G, Schramm G, Rezaei A, Nuyts J, Fulton R, Kyme A, Lois C, Sari H, Price J, Boellaard R, Jeraj R, Bailey DL, Eslick E, Willowson KP, Dutta J. Quantitative PET in the 2020s: a roadmap. Phys Med Biol 2021; 66:06RM01. [PMID: 33339012 PMCID: PMC9358699 DOI: 10.1088/1361-6560/abd4f7] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Positron emission tomography (PET) plays an increasingly important role in research and clinical applications, catalysed by remarkable technical advances and a growing appreciation of the need for reliable, sensitive biomarkers of human function in health and disease. Over the last 30 years, a large amount of the physics and engineering effort in PET has been motivated by the dominant clinical application during that period, oncology. This has led to important developments such as PET/CT, whole-body PET, 3D PET, accelerated statistical image reconstruction, and time-of-flight PET. Despite impressive improvements in image quality as a result of these advances, the emphasis on static, semi-quantitative 'hot spot' imaging for oncologic applications has meant that the capability of PET to quantify biologically relevant parameters based on tracer kinetics has not been fully exploited. More recent advances, such as PET/MR and total-body PET, have opened up the ability to address a vast range of new research questions, from which a future expansion of applications and radiotracers appears highly likely. Many of these new applications and tracers will, at least initially, require quantitative analyses that more fully exploit the exquisite sensitivity of PET and the tracer principle on which it is based. It is also expected that they will require more sophisticated quantitative analysis methods than those that are currently available. At the same time, artificial intelligence is revolutionizing data analysis and impacting the relationship between the statistical quality of the acquired data and the information we can extract from the data. In this roadmap, leaders of the key sub-disciplines of the field identify the challenges and opportunities to be addressed over the next ten years that will enable PET to realise its full quantitative potential, initially in research laboratories and, ultimately, in clinical practice.
Collapse
Affiliation(s)
- Steven R Meikle
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
| | - Vesna Sossi
- Department of Physics and Astronomy, University of British Columbia, Canada
| | - Emilie Roncali
- Department of Biomedical Engineering, University of California, Davis, United States of America
| | - Simon R Cherry
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Richard Banati
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Brain and Mind Centre, The University of Sydney, Australia
- Australian Nuclear Science and Technology Organisation, Sydney, Australia
| | - David Mankoff
- Department of Radiology, University of Pennsylvania, United States of America
| | - Terry Jones
- Department of Radiology, University of California, Davis, United States of America
| | - Michelle James
- Department of Radiology, Molecular Imaging Program at Stanford (MIPS), CA, United States of America
- Department of Neurology and Neurological Sciences, Stanford University, CA, United States of America
| | - Julie Sutcliffe
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Internal Medicine, University of California, Davis, CA, United States of America
| | - Jinsong Ouyang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Yoann Petibon
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Chao Ma
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Suleman Surti
- Department of Radiology, University of Pennsylvania, United States of America
| | - Joel S Karp
- Department of Radiology, University of Pennsylvania, United States of America
| | - Ramsey D Badawi
- Department of Biomedical Engineering, University of California, Davis, United States of America
- Department of Radiology, University of California, Davis, United States of America
| | - Taiga Yamaya
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Go Akamatsu
- National Institute of Radiological Sciences (NIRS), National Institutes for Quantum and Radiological Science and Technology (QST), Chiba, Japan
| | - Georg Schramm
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Ahmadreza Rezaei
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Johan Nuyts
- Department of Imaging and Pathology, Nuclear Medicine & Molecular imaging, KU Leuven, Belgium
| | - Roger Fulton
- Brain and Mind Centre, The University of Sydney, Australia
- Department of Medical Physics, Westmead Hospital, Sydney, Australia
| | - André Kyme
- Brain and Mind Centre, The University of Sydney, Australia
- School of Biomedical Engineering, Faculty of Engineering and IT, The University of Sydney, Australia
| | - Cristina Lois
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, United States of America
| | - Hasan Sari
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Julie Price
- Department of Radiology, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
- Athinoula A. Martinos Center, Massachusetts General Hospital & Harvard Medical School, Boston, MA, United States of America
| | - Ronald Boellaard
- Radiology and Nuclear Medicine, Cancer Center Amsterdam, Amsterdam University Medical Center, location VUMC, Netherlands
| | - Robert Jeraj
- Departments of Medical Physics, Human Oncology and Radiology, University of Wisconsin, United States of America
- Faculty of Mathematics and Physics, University of Ljubljana, Slovenia
| | - Dale L Bailey
- Sydney School of Health Sciences, Faculty of Medicine and Health, The University of Sydney, Australia
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Enid Eslick
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
| | - Kathy P Willowson
- Department of Nuclear Medicine, Royal North Shore Hospital, Sydney, Australia
- Faculty of Science, The University of Sydney, Australia
| | - Joyita Dutta
- Department of Electrical and Computer Engineering, University of Massachusetts Lowell, United States of America
| |
Collapse
|
19
|
Evaluation of Glycolytic Response to Multiple Classes of Anti-glioblastoma Drugs by Noninvasive Measurement of Pyruvate Kinase M2 Using [ 18F]DASA-23. Mol Imaging Biol 2021; 22:124-133. [PMID: 30989436 DOI: 10.1007/s11307-019-01353-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE Pyruvate kinase M2 (PKM2) catalyzes the final step in glycolysis, the key process of tumor metabolism. PKM2 is found in high levels in glioblastoma (GBM) cells with marginal expression within healthy brain tissue, rendering it a key biomarker of GBM metabolic re-programming. Our group has reported the development of a novel radiotracer, 1-((2-fluoro- 6-[18F]fluorophenyl)sulfonyl)-4-((4-methoxyphenyl)sulfonyl)piperazine ([18F]DASA- 23), to non-invasively detect PKM2 levels with positron emission tomography (PET). PROCEDURE U87 human GBM cells were treated with the IC50 concentration of various agents used in the treatment of GBM, including alkylating agents (temozolomide, carmustine, lomustine, procarbazine), inhibitor of topoisomerase I (irinotecan), vascular endothelial and epidermal growth factor receptor inhibitors (cediranib and erlotinib, respectively) anti-metabolite (5-fluorouracil), microtubule inhibitor (vincristine), and metabolic agents (dichloroacetate and IDH1 inhibitor ivosidenib). Following drug exposure for three or 6 days (n = 6 replicates per condition), the radiotracer uptake of [18F]DASA-23 and 2-deoxy-2-[18F]fluoro-D-glucose ([18F]FDG) was assessed. Changes in PKM2 protein levels were determined via Western blot and correlated to radiotracer uptake. RESULTS Significant interactions were found between the treatment agent (n = 12 conditions total comprised 11 drugs and vehicle) and the duration of treatment (3- or 6-day exposure to each drug) on the cellular uptake of [18F]DASA-23 (p = 0.0001). The greatest change in the cellular uptake of [18F]DASA-23 was found after exposure to alkylating agents (p < 0. 0001) followed by irinotecan (p = 0. 0012), erlotinib (p = 0. 02), and 5-fluorouracil (p = 0. 005). Correlation of PKM2 protein levels and [18F]DASA-23 cellular uptake revealed a moderate correlation (r = 0.44, p = 0.15). CONCLUSIONS These proof of principle studies emphasize the superiority of [18F]DASA-23 to [18F]FDG in detecting the glycolytic response of GBM to multiple classes of anti-neoplastic drugs in cell culture. A clinical trial evaluating the diagnostic utility of [18F]DASA-23 PET in GBM patients (NCT03539731) is ongoing.
Collapse
|
20
|
Gelardi F, Kirienko M, Sollini M. Climbing the steps of the evidence-based medicine pyramid: highlights from Annals of Nuclear Medicine 2019. Eur J Nucl Med Mol Imaging 2020; 48:1293-1301. [PMID: 33150459 DOI: 10.1007/s00259-020-05073-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 10/12/2020] [Indexed: 12/17/2022]
Abstract
We aimed to provide an overview on research path in nuclear medicine climbing the steps of the Evidence-Based Medicine (EBM) pyramid using review of 14 subjectively selected papers out of 111 published in the Annals of Nuclear Medicine during January-December 2019. Following the structure of the EBM hierarchy, we chose at least one study for each step of the pyramid from the basis (pre-clinical research, expert opinion, case report and case series), to the middle (case-control and cohort studies, randomised controlled trials), towards the top (meta-analyses and systematic reviews). Additionally, we collected information on the promoter of each included study: investigator-initiated trials (IITs) vs industry-sponsored trials (ISTs). We found that pre-clinical studies are primarily focused on the development of novel molecular targets in cancer, with promising results. At the same time, clinical investigations deal with cardiological, neurological, infectious and oncological applications using both SPECT and PET modalities. Additionally, radionuclide therapy gained interest and is experiencing comprehensive clinical implementation. Our overview confirms the current central role of IITs as compared with ISTs. Challenges and future directions in Nuclear Medicine research are discussed.
Collapse
Affiliation(s)
- Fabrizia Gelardi
- Humanitas University, Pieve Emanuele, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| | - Margarita Kirienko
- Nuclear Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Via Venezian 1, 20133, Milan, Italy.
| | - Martina Sollini
- Humanitas University, Pieve Emanuele, Milan, Italy.,Humanitas Clinical and Research Center - IRCCS, Rozzano, Milan, Italy
| |
Collapse
|
21
|
Garrow AA, Andrews JPM, Gonzalez ZN, Corral CA, Portal C, Morgan TEF, Walton T, Wilson I, Newby DE, Lucatelli C, Tavares AAS. Preclinical dosimetry models and the prediction of clinical doses of novel positron emission tomography radiotracers. Sci Rep 2020; 10:15985. [PMID: 32994530 PMCID: PMC7525662 DOI: 10.1038/s41598-020-72830-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Accepted: 09/07/2020] [Indexed: 11/09/2022] Open
Abstract
Dosimetry models using preclinical positron emission tomography (PET) data are commonly employed to predict the clinical radiological safety of novel radiotracers. However, unbiased clinical safety profiling remains difficult during the translational exercise from preclinical research to first-in-human studies for novel PET radiotracers. In this study, we assessed PET dosimetry data of six 18F-labelled radiotracers using preclinical dosimetry models, different reconstruction methods and quantified the biases of these predictions relative to measured clinical doses to ease translation of new PET radiotracers to first-in-human studies. Whole-body PET images were taken from rats over 240 min after intravenous radiotracer bolus injection. Four existing and two novel PET radiotracers were investigated: [18F]FDG, [18F]AlF-NOTA-RGDfK, [18F]AlF-NOTA-octreotide ([18F]AlF-NOTA-OC), [18F]AlF-NOTA-NOC, [18F]ENC2015 and [18F]ENC2018. Filtered-back projection (FBP) and iterative methods were used for reconstruction of PET data. Predicted and true clinical absorbed doses for [18F]FDG and [18F]AlF-NOTA-OC were then used to quantify bias of preclinical model predictions versus clinical measurements. Our results show that most dosimetry models were biased in their predicted clinical dosimetry compared to empirical values. Therefore, normalization of rat:human organ sizes and correction for reconstruction method biases are required to achieve higher precision of dosimetry estimates.
Collapse
Affiliation(s)
- Adam A Garrow
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Jack P M Andrews
- University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Zaniah N Gonzalez
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Carlos A Corral
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Christophe Portal
- Edinburgh Molecular Imaging (EMI), Nine Edinburgh Bioquarter, Edinburgh, EH16 4UX, UK
| | - Timaeus E F Morgan
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Tashfeen Walton
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Ian Wilson
- Edinburgh Molecular Imaging (EMI), Nine Edinburgh Bioquarter, Edinburgh, EH16 4UX, UK
| | - David E Newby
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.,University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK
| | - Christophe Lucatelli
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Adriana A S Tavares
- Preclinical PET-CT Facility, Edinburgh Imaging, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK. .,University/BHF Centre for Cardiovascular Science, Queen's Medical Research Institute, Edinburgh, EH16 4TJ, UK.
| |
Collapse
|
22
|
Vass L, Fisk M, Lee S, Wilson FJ, Cheriyan J, Wilkinson I. Advances in PET to assess pulmonary inflammation: A systematic review. Eur J Radiol 2020; 130:109182. [PMID: 32702614 DOI: 10.1016/j.ejrad.2020.109182] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 05/27/2020] [Accepted: 07/07/2020] [Indexed: 12/12/2022]
|
23
|
Status of Brain Imaging in Gastroparesis. GASTROINTESTINAL DISORDERS 2020. [DOI: 10.3390/gidisord2020006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
The pathophysiology of nausea and vomiting in gastroparesis is complicated and multifaceted involving the collaboration of both the peripheral and central nervous systems. Most treatment strategies and studies performed in gastroparesis have focused largely on the peripheral effects of this disease, while our understanding of the central nervous system mechanisms of nausea in this entity is still evolving. The ability to view the brain with different neuroimaging techniques has enabled significant advances in our understanding of the central emetic reflex response. However, not enough studies have been performed to further explore the brain–gut mechanisms involved in nausea and vomiting in patients with gastroparesis. The purpose of this review article is to assess the current status of brain imaging and summarize the theories about our present understanding on the central mechanisms involved in nausea and vomiting (N/V) in patients with gastroparesis. Gaining a better understanding of the complex brain circuits involved in the pathogenesis of gastroparesis will allow for the development of better antiemetic prophylactic and treatment strategies.
Collapse
|
24
|
Altine B, Gai Y, Han N, Jiang Y, Ji H, Fang H, Niyonkuru A, Bakari KH, Rajab Arnous MM, Liu Q, Zhang Y, Lan X. Preclinical Evaluation of a Fluorine-18 ( 18F)-Labeled Phosphatidylinositol 3-Kinase Inhibitor for Breast Cancer Imaging. Mol Pharm 2019; 16:4563-4571. [PMID: 31553879 DOI: 10.1021/acs.molpharmaceut.9b00690] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Abstract
Breast cancer is one of the commonest malignancies in women, especially in middle-aged and elderly women. Abnormal activation of the phosphatidylinositol 3-kinase/protein kinase B/mammalian target of rapamycin (PI3K/AKt/mTOR) pathway has been found to be involved in breast cancer proliferation. Pictilisib (GDC-0941) is a potent inhibitor of PI3K with high affinity and is undergoing phase 2 clinical trials. In this study, we aimed to develop a noninvasive PI3K radiotracer to help determine the mechanism of the PI3K/AKt/mTOR pathway to aid in diagnosis. We designed a new 18F-radiolabeled radiotracer based on the structure of pictilisib, to evaluate noninvasively abnormal activation of the PI3K/AKT/mTOR pathway. To increase the water solubility, and to decrease hepatobiliary and gastrointestinal uptake of the tracer, pictilisib was modified with triethylene glycol di(p-toluenesulfonate) (TsO-PEG3-OTs) to obtain TsO-PEG3-GDC-0941 as the precursor for 18F labeling. A nonradiolabeled reference compound [19F]-PEG3-GDC-0941 was also prepared. Breast cancer cell lines, MCF-7 and MDA-MB-231, were used as high- and low-expression PI3K models, respectively. PET imaging and ex vivo biodistribution assays of [18F]-PEG3-GDC-0941 in MCF-7 and MDA-MB-231 xenografts were also performed, and the results were compared. The precursor compound and reference standard compound were successfully synthesized and identified using NMR and mass spectroscopy. The 18F radiolabeling was achieved with a high yield (61 ± 1%) at a high molar activity (2100 ± 100 MBq/mg). MicroPET images and biodistribution studies showed a higher uptake of the radiotracer in MCF-7 tumors than in MDA-MB-231 tumors (7.56 ± 1.01%ID/g vs 4.07 ± 0.68%ID/g, 1 h postinjection). Additionally, the MCF-7 tumor uptake was significantly decreased when a blocking dose of GDC-0941 was coinjected, indicating high specificity. The liver was found to be the major excretory organ with 5.82 ± 0.88%ID/g at 30 min postinjection for MCF-7 mice. This radiotracer holds great potential for patient screening, diagnosis, and therapy prediction of PI3K-related diseases.
Collapse
Affiliation(s)
- Bouhari Altine
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Yongkang Gai
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Na Han
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Yaqun Jiang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Hao Ji
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Hanyi Fang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Alexandre Niyonkuru
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Khamis Hassan Bakari
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Maher Mohamad Rajab Arnous
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Qingyao Liu
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Yongxue Zhang
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| | - Xiaoli Lan
- Department of Nuclear Medicine, Union Hospital, Tongji Medical College , Huazhong University of Science and Technology , Wuhan 430022 , China.,Hubei Province Key Laboratory of Molecular Imaging , Wuhan 430022 , China
| |
Collapse
|
25
|
Radaram B, Pisaneschi F, Rao Y, Yang P, Piwnica-Worms D, Alauddin MM. Novel derivatives of anaplastic lymphoma kinase inhibitors: Synthesis, radiolabeling, and preliminary biological studies of fluoroethyl analogues of crizotinib, alectinib, and ceritinib. Eur J Med Chem 2019; 182:111571. [PMID: 31425908 DOI: 10.1016/j.ejmech.2019.111571] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/13/2022]
Abstract
Anaplastic lymphoma kinase (ALK), an oncogenic receptor tyrosine kinase, is a therapeutic target in various cancers, including non-small cell lung cancer. Although several ALK inhibitors, including crizotinib, ceritinib, and alectinib, are approved for cancer treatment, their long-term benefit is often limited by the cancer's acquisition of resistance owing to secondary point mutations in ALK. Importantly, some ALK inhibitors cannot cross the blood-brain barrier (BBB) and thus have little or no efficacy against brain metastases. The introduction of a lipophilic moiety, such as a fluoroethyl group may improve the drug's BBB penetration. Herein, we report the synthesis of fluoroethyl analogues of crizotinib 1, alectinib 4, and ceritinib 9, and their radiolabeling with 18F for pharmacokinetic studies. The fluoroethyl derivatives and their radioactive analogues were obtained in good yields with high purity and good molar activity. A cytotoxicity screen in ALK-expressing H2228 lung cancer cells showed that the analogues had up to nanomolar potency and the addition of the fluorinated moiety had minimal impact overall on the potency of the original drugs. Positron emission tomography in healthy mice showed that the analogues had enhanced BBB penetration, suggesting that they have therapeutic potential against central nervous system metastases.
Collapse
Affiliation(s)
- Bhasker Radaram
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Federica Pisaneschi
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yi Rao
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Ping Yang
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - David Piwnica-Worms
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Mian M Alauddin
- Department of Cancer Systems Imaging, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
26
|
Jiemy WF, Heeringa P, Kamps JA, van der Laken CJ, Slart RH, Brouwer E. Positron emission tomography (PET) and single photon emission computed tomography (SPECT) imaging of macrophages in large vessel vasculitis: Current status and future prospects. Autoimmun Rev 2018; 17:715-726. [PMID: 29729443 DOI: 10.1016/j.autrev.2018.02.006] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2018] [Accepted: 02/07/2018] [Indexed: 12/21/2022]
|
27
|
Tegnebratt T, Lu L, Eksborg S, Chireh A, Damberg P, Nikkhou-Aski S, Foukakis T, Rundqvist H, Holmin S, Kuiper RV, Samen E. Treatment response assessment with (R)-[ 11CPAQ PET in the MMTV-PyMT mouse model of breast cancer. EJNMMI Res 2018; 8:25. [PMID: 29616369 PMCID: PMC5882477 DOI: 10.1186/s13550-018-0380-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 03/23/2018] [Indexed: 02/07/2023] Open
Abstract
Background The goal of the study was to assess the potential of the vascular endothelial growth factor receptor (VEGFR)-2-targeting carbon-11 labeled (R)-N-(4-bromo-2-fluorophenyl)-6-methoxy-7-((1-methyl-3-piperidinyl)methoxy)-4-quinazolineamine ((R)-[11C]PAQ) as a positron emission tomography (PET) imaging biomarker for evaluation of the efficacy of anticancer drugs in preclinical models. Methods MMTV-PyMT mice were treated with vehicle alone (VEH), murine anti-VEGFA antibody (B20-4.1.1), and paclitaxel (PTX) in combination or as single agents. The treatment response was measured with (R)-[11C]PAQ PET as standardized uptake value (SUV)mean, SUVmax relative changes at the baseline (day 0) and follow-up (day 4) time points, and magnetic resonance imaging (MRI)-derived PyMT mammary tumor volume (TV) changes. Expression of Ki67, VEGFR-2, and CD31 in tumor tissue was determined by immunohistochemistry (IHC). Non-parametric statistical tests were used to evaluate the relation between (R)-[11C]PAQ radiotracer uptake and therapy response biomarkers. Results The (R)-[11C]PAQ SUVmax in tumors was significantly reduced after 4 days in the B20-4.1.1/PTX combinational and B20-4.1.1 monotherapy groups (p < 0.0005 and p < 0.003, respectively). No significant change was observed in the PTX monotherapy group. There was a significant difference in the SUVmax change between the VEH group and B20-4.1.1/PTX combinational group, as well as between the VEH group and the B20-4.1.1 monotherapy group (p < 0.05). MRI revealed significant decreases in TV in the B20-4.1.1/PTX treatment group (p < 0.005) but not the other therapy groups. A positive trend was observed between the (R)-[11C]PAQ SUVmax change and TV reduction in the B20-4.1.1/PTX group. Statistical testing showed a significant difference in the blood vessel density between the B20-4.1.1/PTX combinational group and the VEH group (p < 0.05) but no significant difference in the Ki67 positive signal between treatment groups. Conclusions The results of this study are promising. However, additional studies are necessary before (R)-[11C]PAQ can be approved as a predictive radiotracer for cancer therapy response.
Collapse
Affiliation(s)
- T Tegnebratt
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden. .,Department of Neuroradiology, Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-17176, Stockholm, Sweden.
| | - L Lu
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.,Department of Comparative Medicine, Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - S Eksborg
- Department of Women's and Children's Health, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - A Chireh
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - P Damberg
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.,Department of Comparative Medicine, Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - S Nikkhou-Aski
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.,Department of Comparative Medicine, Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - T Foukakis
- Department of Oncology-Pathology, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - H Rundqvist
- Department of Cell and Molecular Biology, Karolinska Institutet, SE-17176, Stockholm, Sweden
| | - S Holmin
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.,Department of Neuroradiology, Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| | - R V Kuiper
- Core Facility for Morphologic Phenotype Analysis, Laboratory Medicine, Karolinska Institutet, SE-14183, Huddinge, Sweden
| | - E Samen
- Department of Clinical Neuroscience, Karolinska Institutet, SE-17176, Stockholm, Sweden.,Department of Neuroradiology, Karolinska Experimental Research and Imaging Center, Karolinska University Hospital, SE-17176, Stockholm, Sweden
| |
Collapse
|
28
|
Nodwell MB, Yang H, Čolović M, Yuan Z, Merkens H, Martin RE, Bénard F, Schaffer P, Britton R. 18F-Fluorination of Unactivated C-H Bonds in Branched Aliphatic Amino Acids: Direct Synthesis of Oncological Positron Emission Tomography Imaging Agents. J Am Chem Soc 2017; 139:3595-3598. [PMID: 28248493 DOI: 10.1021/jacs.6b11533] [Citation(s) in RCA: 92] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
A mild and selective photocatalytic C-H 18F-fluorination reaction has been developed that provides direct access to 18F-fluorinated amino acids. The biodistribution and uptake of three 18F-labeled leucine analogues via LAT1 mediated transport in several cancer cell lines is reported. Positron emission tomography imaging of mice bearing PC3 (prostate) or U87 (glioma) xenografts using 5-[18F]-fluorohomoleucine showed high tumor uptake and excellent tumor visualization, highlighting the utility of this strategy for rapid tracer discovery for oncology.
Collapse
Affiliation(s)
- Matthew B Nodwell
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada
| | - Hua Yang
- Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Milena Čolović
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Zheliang Yuan
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada.,Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Helen Merkens
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Rainer E Martin
- Medicinal Chemistry, Roche Pharma Research and Early Development (pRED), Roche Innovation Center Basel, F. Hoffmann-La Roche Ltd , Grenzacherstrasse 124, CH-4070 Basel, Switzerland
| | - François Bénard
- Department of Molecular Oncology, BC Cancer Agency , Vancouver, British Columbia V5Z 1L3 Canada
| | - Paul Schaffer
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada.,Life Sciences Division, TRIUMF , Vancouver, British Columbia V6T 2A3, Canada
| | - Robert Britton
- Department of Chemistry, Simon Fraser University , Burnaby, British Columbia V5A 1S2, Canada
| |
Collapse
|
29
|
Opacic T, Paefgen V, Lammers T, Kiessling F. Status and trends in the development of clinical diagnostic agents. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [DOI: 10.1002/wnan.1441] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2016] [Revised: 09/02/2016] [Accepted: 09/15/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Tatjana Opacic
- Department of Experimental Molecular Imaging; RWTH Aachen University; Aachen Germany
| | - Vera Paefgen
- Department of Experimental Molecular Imaging; RWTH Aachen University; Aachen Germany
| | - Twan Lammers
- Department of Experimental Molecular Imaging; RWTH Aachen University; Aachen Germany
- Department of Pharmaceutics; Utrecht University; Utrecht The Netherlands
- Department of Targeted Therapeutics; University of Twente; Enschede The Netherlands
| | - Fabian Kiessling
- Department of Experimental Molecular Imaging; RWTH Aachen University; Aachen Germany
| |
Collapse
|
30
|
Hoogenboom TC, Thursz M, Aboagye EO, Sharma R. Functional imaging of hepatocellular carcinoma. Hepat Oncol 2016; 3:137-153. [PMID: 30191034 DOI: 10.2217/hep-2015-0005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Accepted: 01/20/2016] [Indexed: 02/06/2023] Open
Abstract
Imaging plays a key role in the clinical management of hepatocellular carcinoma (HCC), but conventional imaging techniques have limited sensitivity in visualizing small tumors and assessing response to locoregional treatments and sorafenib. Functional imaging techniques allow visualization of organ and tumor physiology. Assessment of functional characteristics of tissue, such as metabolism, proliferation and stiffness, may overcome some of the limitations of structural imaging. In particular, novel molecular imaging agents offer a potential tool for early diagnosis of HCC, and radiomics may aid in response assessment and generate prognostic models. Further prospective research is warranted to evaluate emerging techniques and their cost-effectiveness in the context of HCC in order to improve detection and response assessment.
Collapse
Affiliation(s)
- Tim Ch Hoogenboom
- Department of Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0HS, UK.,Department of Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0HS, UK
| | - Mark Thursz
- Department of Hepatology, Imperial College NHS Trust, 10th Floor, Norfolk Place, St Mary's Hospital, London, UK.,Department of Hepatology, Imperial College NHS Trust, 10th Floor, Norfolk Place, St Mary's Hospital, London, UK
| | - Eric O Aboagye
- Comprehensive Cancer Imaging Centre at Imperial College, Faculty of Medicine, Imperial College London, GN1, Ground Floor, Commonwealth building, Hammersmith Campus, London, UK.,Comprehensive Cancer Imaging Centre at Imperial College, Faculty of Medicine, Imperial College London, GN1, Ground Floor, Commonwealth building, Hammersmith Campus, London, UK
| | - Rohini Sharma
- Department of Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0HS, UK.,Department of Experimental Medicine, Imperial College London, Hammersmith Hospital, Du Cane Road, London, W12 0HS, UK
| |
Collapse
|
31
|
Recent Advances in the Development and Application of Radiolabeled Kinase Inhibitors for PET Imaging. Molecules 2015; 20:22000-27. [PMID: 26690113 PMCID: PMC6332294 DOI: 10.3390/molecules201219816] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2015] [Revised: 11/18/2015] [Accepted: 12/01/2015] [Indexed: 12/20/2022] Open
Abstract
Over the last 20 years, intensive investigation and multiple clinical successes targeting protein kinases, mostly for cancer treatment, have identified small molecule kinase inhibitors as a prominent therapeutic class. In the course of those investigations, radiolabeled kinase inhibitors for positron emission tomography (PET) imaging have been synthesized and evaluated as diagnostic imaging probes for cancer characterization. Given that inhibitor coverage of the kinome is continuously expanding, in vivo PET imaging will likely find increasing applications for therapy monitoring and receptor density studies both in- and outside of oncological conditions. Early investigated radiolabeled inhibitors, which are mostly based on clinically approved tyrosine kinase inhibitor (TKI) isotopologues, have now entered clinical trials. Novel radioligands for cancer and PET neuroimaging originating from novel but relevant target kinases are currently being explored in preclinical studies. This article reviews the literature involving radiotracer design, radiochemistry approaches, biological tracer evaluation and nuclear imaging results of radiolabeled kinase inhibitors for PET reported between 2010 and mid-2015. Aspects regarding the usefulness of pursuing selective vs. promiscuous inhibitor scaffolds and the inherent challenges associated with intracellular enzyme imaging will be discussed.
Collapse
|
32
|
Dubash SR, Idowu OA, Sharma R. The emerging role of positron emission tomography in hepatocellular carcinoma. Hepat Oncol 2015; 2:191-200. [PMID: 30190998 DOI: 10.2217/hep.15.6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality worldwide. HCC a heterogeneous disease occurring on the background of cirrhosis. The presence of cirrhosis limits the sensitivity of conventional imaging modalities in differentiating HCC from surrounding cirrhotic parenchyma. Positron emission tomography (PET) using 18F-fluorodeoxyglucose (18F-FDG) is widely used for assessing a variety of malignancies, however, has poor sensitivity in the evaluation of HCC. This has led to the investigation of other radiotracers such as 11C-acetate and 11C-choline, with improved sensitivity in terms of detection and therapeutic response. In this review, we discuss the emerging field of PET imaging for the detection, staging and assessment of treatment response in HCC. In particular we discuss the role of 18F-FDG-PET in imaging hepatocellular cancer, the limitations of this PET tracer and emerging novel PET tracers being investigated that exploit key metabolic processes including fatty acid and lipid synthesis, choline kinase activity and gene expression.
Collapse
Affiliation(s)
- Suraiya R Dubash
- Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, UK.,Department of Surgery & Cancer, Imperial College London, Hammersmith Campus, Du Cane Road, London, UK
| | - Oluwagbemiga A Idowu
- Department of Medical Oncology, Hammersmith Hospital, London, UK.,Department of Medical Oncology, Hammersmith Hospital, London, UK
| | - Rohini Sharma
- Division of Translational & Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, UK.,Medical Oncology & Clinical Pharmacology, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK.,Division of Translational & Experimental Medicine, Imperial College London, Hammersmith Campus, Du Cane Road, London, UK.,Medical Oncology & Clinical Pharmacology, Imperial College London, Hammersmith Campus, Du Cane Road, London, W12 0HS, UK
| |
Collapse
|
33
|
Mammatas LH, Verheul HMW, Hendrikse NH, Yaqub M, Lammertsma AA, Menke-van der Houven van Oordt CW. Molecular imaging of targeted therapies with positron emission tomography: the visualization of personalized cancer care. Cell Oncol (Dordr) 2014; 38:49-64. [PMID: 25248503 DOI: 10.1007/s13402-014-0194-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/20/2014] [Indexed: 01/19/2023] Open
Abstract
INTRODUCTION Molecular imaging has been defined as the visualization, characterization and measurement of biological processes at the molecular and cellular level in humans and other living systems. In oncology it enables to visualize (part of) the functional behaviour of tumour cells, in contrast to anatomical imaging that focuses on the size and location of malignant lesions. Available molecular imaging techniques include single photon emission computed tomography (SPECT), positron emission tomography (PET) and optical imaging. In PET, a radiotracer consisting of a positron emitting radionuclide attached to the biologically active molecule of interest is administrated to the patient. Several approaches have been undertaken to use PET for the improvement of personalized cancer care. For example, a variety of radiolabelled ligands have been investigated for intratumoural target identification and radiolabelled drugs have been developed for direct visualization of the biodistibution in vivo, including intratumoural therapy uptake. First indications of the clinical value of PET for target identification and response prediction in oncology have been reported. This new imaging approach is rapidly developing, but uniformity of scanning processes, standardized methods for outcome evaluation and implementation in daily clinical practice are still in progress. In this review we discuss the available literature on molecular imaging with PET for personalized targeted treatment strategies. CONCLUSION Molecular imaging with radiolabelled targeted anticancer drugs has great potential for the improvement of personalized cancer care. The non-invasive quantification of drug accumulation in tumours and normal tissues provides understanding of the biodistribution in relation to therapeutic and toxic effects.
Collapse
Affiliation(s)
- Lemonitsa H Mammatas
- Dept of Medical Oncology VUmc Cancer Center Amsterdam, VU University Medical Center, De Boelelaan 1117, 1081 HV, Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
34
|
Sieren JC, Meyerholz DK, Wang XJ, Davis BT, Newell JD, Hammond E, Rohret JA, Rohret FA, Struzynski JT, Goeken JA, Naumann PW, Leidinger MR, Taghiyev A, Van Rheeden R, Hagen J, Darbro BW, Quelle DE, Rogers CS. Development and translational imaging of a TP53 porcine tumorigenesis model. J Clin Invest 2014; 124:4052-66. [PMID: 25105366 DOI: 10.1172/jci75447] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2014] [Accepted: 06/19/2014] [Indexed: 01/03/2023] Open
Abstract
Cancer is the second deadliest disease in the United States, necessitating improvements in tumor diagnosis and treatment. Current model systems of cancer are informative, but translating promising imaging approaches and therapies to clinical practice has been challenging. In particular, the lack of a large-animal model that accurately mimics human cancer has been a major barrier to the development of effective diagnostic tools along with surgical and therapeutic interventions. Here, we developed a genetically modified porcine model of cancer in which animals express a mutation in TP53 (which encodes p53) that is orthologous to one commonly found in humans (R175H in people, R167H in pigs). TP53(R167H/R167H) mutant pigs primarily developed lymphomas and osteogenic tumors, recapitulating the tumor types observed in mice and humans expressing orthologous TP53 mutant alleles. CT and MRI imaging data effectively detected developing tumors, which were validated by histopathological evaluation after necropsy. Molecular genetic analyses confirmed that these animals expressed the R167H mutant p53, and evaluation of tumors revealed characteristic chromosomal instability. Together, these results demonstrated that TP53(R167H/R167H) pigs represent a large-animal tumor model that replicates the human condition. Our data further suggest that this model will be uniquely suited for developing clinically relevant, noninvasive imaging approaches to facilitate earlier detection, diagnosis, and treatment of human cancers.
Collapse
|
35
|
Dall'Angelo S, Bandaranayaka N, Windhorst AD, Vugts DJ, van der Born D, Onega M, Schweiger LF, Zanda M, O'Hagan D. Tumour imaging by Positron Emission Tomography using fluorinase generated 5-[18F]fluoro-5-deoxyribose as a novel tracer. Nucl Med Biol 2013; 40:464-70. [DOI: 10.1016/j.nucmedbio.2013.02.006] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Revised: 02/08/2013] [Accepted: 02/13/2013] [Indexed: 11/29/2022]
|
36
|
Smith G, Carroll L, Aboagye EO. New frontiers in the design and synthesis of imaging probes for PET oncology: current challenges and future directions. Mol Imaging Biol 2013; 14:653-66. [PMID: 22948535 DOI: 10.1007/s11307-012-0590-y] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Despite being developed over 30 years ago, 2-deoxy-2-[(18)F]fluoro-D-glucose remains the most frequently used radiotracer in PET oncology. In the last decade, interest in new and more specific radiotracers for imaging biological processes of oncologic interest has increased exponentially. This review summarizes the strategies underlying the development of those probes together with their validation and status of clinical translation; a brief summary of new radiochemistry strategies applicable to PET imaging is also included. The article finishes with a consideration of the challenges imaging scientists must overcome to bring about increased adoption of PET as a diagnostic or pharmacologic tool.
Collapse
Affiliation(s)
- Graham Smith
- Post-Graduate Medical Institute, University of Hull, Cottingham Road, Hull, HU6 7RX, UK
| | | | | |
Collapse
|
37
|
Pinato DJ, Stavraka C, Tanner M, Esson A, Jacobson EW, Wilkins MR, Libri V. Clinical, ethical and financial implications of incidental imaging findings: experience from a phase I trial in healthy elderly volunteers. PLoS One 2012; 7:e49814. [PMID: 23166776 PMCID: PMC3500322 DOI: 10.1371/journal.pone.0049814] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2012] [Accepted: 10/17/2012] [Indexed: 12/05/2022] Open
Abstract
Background The detection of incidental findings (IF) in magnetic resonance imaging (MRI) studies is common and increases as a function of age. Responsible handling of IF is required, with implications for the conduct of research and the provision of good clinical care. Aim To investigate the prevalence and clinical significance of IF in a prospective cohort of healthy elderly volunteers who underwent MRI of the torso as a baseline investigation for a phase I trial. We assessed the follow-up pathway with consequent cost implications and impact on trial outcomes. Methods A total of 29 elderly healthy volunteers (mean age 67, range 61–77, 59% female) were eligible at screening and underwent MRI for assessment of visceral and subcutaneous fat. Results IF were detected in 19 subjects (66%). Suspected IF of high and low clinical significance were found in 14% and 52% of participants, respectively. Follow up of IF was conducted in 18 individuals, confirming abnormalities in 13 subjects, 3 of whom were recommended for deferred clinical re-evaluation. The remaining 5 subjects had false positive IF based on second line imaging tests. Costs of follow-up medical care were considerable. Conclusion MRI abnormalities are common in elderly individuals, as a result of age and non-diagnostic quality of research scans. In the presence of IF in the context of clinical trials, immediate referrals and follow up assessments may be required to rule out suspected pathology prior to exposing trial participants to investigational medicine products (IMP). Unanticipated costs, ethical implication and the possible impact of IF on trial outcomes need to be taken into account when designing and conducting trials with an IMP.
Collapse
Affiliation(s)
- David J. Pinato
- The National Institute for Health Research (NIHR)-Wellcome Trust Imperial College Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Chara Stavraka
- The National Institute for Health Research (NIHR)-Wellcome Trust Imperial College Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Mark Tanner
- Imanova Centre for Imaging Sciences, London, United Kingdom
| | - Audrey Esson
- The National Institute for Health Research (NIHR)-Wellcome Trust Imperial College Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Eric W. Jacobson
- Sirtris Pharmaceuticals Inc., Cambridge, Massachusetts, United States of America
| | - Martin R. Wilkins
- The National Institute for Health Research (NIHR)-Wellcome Trust Imperial College Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
| | - Vincenzo Libri
- The National Institute for Health Research (NIHR)-Wellcome Trust Imperial College Clinical Research Facility, Imperial Centre for Translational and Experimental Medicine, Imperial College, Hammersmith Hospital Campus, London, United Kingdom
- * E-mail:
| |
Collapse
|
38
|
Nguyen QD, Challapalli A, Smith G, Fortt R, Aboagye EO. Imaging apoptosis with positron emission tomography: 'bench to bedside' development of the caspase-3/7 specific radiotracer [(18)F]ICMT-11. Eur J Cancer 2012; 48:432-40. [PMID: 22226480 DOI: 10.1016/j.ejca.2011.11.033] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2011] [Accepted: 11/21/2011] [Indexed: 12/20/2022]
Abstract
The capacity to evade apoptosis has been defined as one of the hallmarks of cancer and, thus, effective anti-cancer therapy often induces apoptosis. A biomarker for imaging apoptosis could assist in monitoring the efficacy of a wide range of current and future therapeutics. Despite the potential, there are limited clinical examples of the use of positron emission tomography for imaging of apoptosis. [(18)F]ICMT-11 is a novel reagent designed to non-invasively image caspase-3 activation and, hence, drug-induced apoptosis. Radiochemistry development of [(18)F]ICMT-11 has been undertaken to improve specific radioactivity, reduce content of stable impurities, reduce synthesis time and enable automation for manufacture of multi-patient dose. Due to the promising mechanistic and safety profile of [(18)F]ICMT-11, the radiotracer is transitioning to clinical development and has been selected as a candidate radiotracer by the QuIC-ConCePT consortium for further evaluation in preclinical models and humans. A successful outcome will allow use of the radiotracer as qualified method for evaluating the pharmaceutical industry's next generation therapeutics.
Collapse
Affiliation(s)
- Quang-Dé Nguyen
- Department of Surgery and Cancer, Imperial College, London, UK
| | | | | | | | | |
Collapse
|
39
|
Lawrence AJ, Heinz A. Imaging - the interface with pharmacology: looking to the future. Br J Pharmacol 2011; 163:1563-4. [DOI: 10.1111/j.1476-5381.2011.01294.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022] Open
|