1
|
Cirak Z, Tanoglu A, Yeniceri M, Tanoglu EG, Kaplan M, Sade AG. Certolizumab Has Favorable Efficacy on Preventing Pancreas and Target Organs Damage in Acute Pancreatitis. Pancreas 2024; 53:e588-e594. [PMID: 38986079 DOI: 10.1097/mpa.0000000000002343] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 07/12/2024]
Abstract
OBJECTIVE It was targeted to assess the efficacy of certolizumab on pancreas and target organs via biochemical parameters and histopathologic scores in experimental acute pancreatitis (AP). MATERIALS AND METHODS Forty male Sprague Dawley rats were divided into the following 5 equal groups: group 1 (sham group), group 2 (AP group), group 3 (AP + low-dose certolizumab group), group 4 (AP + high-dose certolizumab group), and group 5 (placebo group). Rats in all groups were sacrificed 24 hours after the last injection and amylase, tumor necrosis factor α, transforming growth factor β, interleukin 1β, malondialdehyde, superoxide dismutase, and glutathione peroxidase levels were studied in blood samples. Histopathological investigation of both the pancreas and target organs (lungs, liver, heart, kidneys) was performed by a pathologist blind to the groups. In silico analysis were also accomplished. RESULTS The biochemical results in the certolizumab treatment groups were identified to be significantly favorable compared to the AP group (P < 0.001). The difference between the high-dose group (group 4) and low-dose treatment group (group 3) was found to be significant in terms of biochemical parameters and histopathological scores (P < 0.001). In terms of the effect of certolizumab treatment on the target organs (especially on lung tissue), the differences between the low-dose treatment group (group 3) and high-dose treatment group (group 4) with the AP group (group 2) were significant. CONCLUSIONS Certolizumab has favorable protective effects on pancreas and target organs in AP. It may be a beneficial agent for AP treatment and may prevent target organ damage.
Collapse
Affiliation(s)
- Zafer Cirak
- From the Department of Internal Medicine, Honaz State Hospital
| | - Alpaslan Tanoglu
- Department of Internal Medicine, Division of Gastroenterology, Bahçeşehir University, Faculty of Medicine
| | - Murat Yeniceri
- University of Health Sciences, Institution of Hamidiye Health Sciences, Department of Molecular Biology and Genetics
| | - Esra Guzel Tanoglu
- Department of Internal Medicine, University of Health Sciences, Bakırköy Dr. Sadi Konuk Hospital
| | - Mustafa Kaplan
- Department of Internal Medicine, University of Health Sciences, Sultan Abdülhamid Han Hospital
| | - Ayşe Gökcen Sade
- Department of Pathology, University of Health Sciences, Sultan Abdülhamid Han Hospital, Istanbul, Turkey
| |
Collapse
|
2
|
Lucinian YA, Martineau P, Abikhzer G, Harel F, Pelletier-Galarneau M. Novel tracers to assess myocardial inflammation with radionuclide imaging. J Nucl Cardiol 2024:102012. [PMID: 39069249 DOI: 10.1016/j.nuclcard.2024.102012] [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: 02/16/2024] [Revised: 07/08/2024] [Accepted: 07/15/2024] [Indexed: 07/30/2024]
Abstract
Myocardial inflammation plays a central role in the pathophysiology of various cardiac diseases. While FDG-PET is currently the primary method for molecular imaging of myocardial inflammation, its effectiveness is hindered by physiological myocardial uptake as well as its propensity for uptake by multiple disease-specific mechanisms. Novel radiotracers targeting diverse inflammatory immune cells and molecular pathways may provide unique insight through the visualization of underlying mechanisms central to the pathogenesis of inflammatory cardiac diseases, offering opportunities for increased understanding of immunocardiology. Moreover, the potentially enhanced specificity may lead to better quantification of disease activity, aiding in the guidance and monitoring of immunomodulatory therapy. This review aims to provide an update on advancements in non-FDG radiotracers for imaging myocardial inflammatory diseases, with a focus on cardiac sarcoidosis, myocarditis, and acute myocardial infarction.
Collapse
Affiliation(s)
| | | | - Gad Abikhzer
- Jewish General Hospital, Montreal, Quebec, Canada
| | | | | |
Collapse
|
3
|
Yan G, Wang X, Fan Y, Lin J, Yan J, Wang L, Pan D, Xu Y, Yang M. Immuno-PET Imaging of TNF-α in Colitis Using 89Zr-DFO-infliximab. Mol Pharm 2022; 19:3632-3639. [PMID: 36039398 DOI: 10.1021/acs.molpharmaceut.2c00411] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Tumor necrosis factor-alpha (TNF-α) neutralization has become increasingly important in the treatment of inflammatory bowel diseases (IBD). A series of monoclonal antibodies were approved in the clinic for anti-TNF-α therapy. However, a comprehensive assessment of TNF-α levels throughout the colon, which facilitates the diagnosis of IBD and predicts anti-TNF-α efficacy, remains challenging. Here, we radiolabeled infliximab with long-lived radionuclides 89Zr for immuno-positron emission tomography (PET) imaging of TNF-α in vivo. The increased TNF-α level was detected in the inflammatory colon of the dextran sodium sulfate-induced colitis mice. The immuno-PET imaging of 89Zr-desferrioxamine-infliximab reveals a high uptake (7.1 ± 0.3%ID/g) in the inflammatory colon, which is significantly higher than in the healthy control and blocked groups. The colon-to-muscle ratio reached more than 10 and was maintained at a high level for 10 h after injection. The ex vivo biodistribution study also verified the superior uptake in the inflammatory colon. This study provides an in vivo immune-PET approach to molecular imaging of the pro-inflammatory cytokine TNF-α. It is promising in diagnosing and predicting efficacy in both IBD and other autoimmune diseases.
Collapse
Affiliation(s)
- Ge Yan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.,NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Xinyu Wang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.,NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Yeli Fan
- College of Environmental Engineering, Wuxi University, Wuxi 214105, PR China
| | - Jianhan Lin
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.,NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Junjie Yan
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.,NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Lizhen Wang
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Donghui Pan
- NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Yuping Xu
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.,NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| | - Min Yang
- School of Pharmacy, Nanjing Medical University, Nanjing 211166, PR China.,NHC Key Laboratory of Nuclear Medicine, Jiangsu Key Laboratory of Molecular Nuclear Medicine, Jiangsu Institute of Nuclear Medicine, Wuxi 214063, PR China
| |
Collapse
|
4
|
Navarro-Compán V, Ermann J, Poddubnyy D. A glance into the future of diagnosis and treatment of spondyloarthritis. Ther Adv Musculoskelet Dis 2022; 14:1759720X221111611. [PMID: 35898564 PMCID: PMC9310200 DOI: 10.1177/1759720x221111611] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/18/2022] [Indexed: 11/16/2022] Open
Abstract
The last two decades have seen major developments in the field of spondyloarthritis (SpA), but there are still important unmet needs to address. In the future, we envisage important advances in the diagnosis and treatment of SpA. In the diagnosis of SpA, the use of online and social media tools will increase awareness of the disease and facilitate the referral of patients to rheumatology clinics. In addition, more specific diagnostic tests will be available, especially advanced imaging methods and new biomarkers. This will allow most patients to be diagnosed at an early stage of the disease. In the treatment of SpA, an increasing number of novel treatment targets can be expected, most of which will be directed against intracellular enzymes. We hope to see more strategy trials shaping treatment pathways in SpA and accommodating principals of precision medicine. Approved treatment options will be available for both axial and peripheral SpA. We also hope to intervene not only at the inflammation level but also at the level of underlying immunological processes that might be associated with a higher probability of long-standing remission if not a cure. Finally, artificial intelligence techniques will allow for the analysis of large-scale data to answer relevant research questions for the diagnosis and management of patients with SpA.
Collapse
Affiliation(s)
| | - Joerg Ermann
- Division of Rheumatology, Inflammation and
Immunity, Brigham and Women’s Hospital and Harvard Medical School, Boston,
MA, USA
| | - Denis Poddubnyy
- Department of Gastroenterology, Infectiology
and Rheumatology (Including Nutrition Medicine), Charité –
Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and
Humboldt-Universität zu Berlin, Hindenburgdamm 30, Berlin 12203,
Germany
- Epidemiology Unit, German Rheumatism Research
Centre, Berlin, Germany
| |
Collapse
|
5
|
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: 8] [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
|
6
|
Rösner L, Konken CP, Depke DA, Rentmeister A, Schäfers M. Covalent labeling of immune cells. Curr Opin Chem Biol 2022; 68:102144. [DOI: 10.1016/j.cbpa.2022.102144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Revised: 03/06/2022] [Accepted: 03/11/2022] [Indexed: 12/15/2022]
|
7
|
Beckford-Vera DR, Flavell RR, Seo Y, Martinez-Ortiz E, Aslam M, Thanh C, Fehrman E, Pardons M, Kumar S, Deitchman AN, Ravanfar V, Schulte B, Wu IWK, Pan T, Reeves JD, Nixon CC, Iyer NS, Torres L, Munter SE, Hyunh T, Petropoulos CJ, Hoh R, Franc BL, Gama L, Koup RA, Mascola JR, Chomont N, Deeks SG, VanBrocklin HF, Henrich TJ. First-in-human immunoPET imaging of HIV-1 infection using 89Zr-labeled VRC01 broadly neutralizing antibody. Nat Commun 2022; 13:1219. [PMID: 35264559 PMCID: PMC8907355 DOI: 10.1038/s41467-022-28727-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 02/01/2022] [Indexed: 11/09/2022] Open
Abstract
A major obstacle to achieving long-term antiretroviral (ART) free remission or functional cure of HIV infection is the presence of persistently infected cells that establish a long-lived viral reservoir. HIV largely resides in anatomical regions that are inaccessible to routine sampling, however, and non-invasive methods to understand the longitudinal tissue-wide burden of HIV persistence are urgently needed. Positron emission tomography (PET) imaging is a promising strategy to identify and characterize the tissue-wide burden of HIV. Here, we assess the efficacy of using immunoPET imaging to characterize HIV reservoirs and identify anatomical foci of persistent viral transcriptional activity using a radiolabeled HIV Env-specific broadly neutralizing antibody, 89Zr-VRC01, in HIV-infected individuals with detectable viremia and on suppressive ART compared to uninfected controls (NCT03729752). We also assess the relationship between PET tracer uptake in tissues and timing of ART initiation and direct HIV protein expression in CD4 T cells obtained from lymph node biopsies. We observe significant increases in 89Zr-VRC01 uptake in various tissues (including lymph nodes and gut) in HIV-infected individuals with detectable viremia (N = 5) and on suppressive ART (N = 5) compared to uninfected controls (N = 5). Importantly, PET tracer uptake in inguinal lymph nodes in viremic and ART-suppressed participants significantly and positively correlates with HIV protein expression measured directly in tissue. Our strategy may allow non-invasive longitudinal characterization of residual HIV infection and lays the framework for the development of immunoPET imaging in a variety of other infectious diseases.
Collapse
Affiliation(s)
- Denis R Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Enrique Martinez-Ortiz
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Maya Aslam
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Cassandra Thanh
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Emily Fehrman
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Marion Pardons
- Department of Microbiology, Infectiology and Immunology, Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Shreya Kumar
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Amelia N Deitchman
- Department of Clinical Pharmacy, University of California, San Francisco, USA
| | - Vahid Ravanfar
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Brailee Schulte
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - I-Wei Katherine Wu
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Tony Pan
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Jacqueline D Reeves
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Christopher C Nixon
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Nikita S Iyer
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Leonel Torres
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Sadie E Munter
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Tony Hyunh
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Christos J Petropoulos
- Monogram Biosciences, Inc., Laboratory Corporation of America, South San Francisco, San Francisco, USA
| | - Rebecca Hoh
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Benjamin L Franc
- Department of Radiology, Stanford University, Palo Alto, CA, USA
| | - Lucio Gama
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Richard A Koup
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - John R Mascola
- Vaccine Research Center, National Institute for Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Nicolas Chomont
- Department of Microbiology, Infectiology and Immunology, Centre de Recherche du CHUM, Université de Montréal, Montreal, QC, Canada
| | - Steven G Deeks
- Division of HIV, Infectious Diseases and Global Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Henry F VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA.
| | - Timothy J Henrich
- Division of Experimental Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
8
|
Aarntzen EHJG, Noriega-Álvarez E, Artiko V, Dias AH, Gheysens O, Glaudemans AWJM, Lauri C, Treglia G, van den Wyngaert T, van Leeuwen FWB, Terry SYA. EANM recommendations based on systematic analysis of small animal radionuclide imaging in inflammatory musculoskeletal diseases. EJNMMI Res 2021; 11:85. [PMID: 34487263 PMCID: PMC8421483 DOI: 10.1186/s13550-021-00820-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/02/2021] [Indexed: 11/26/2022] Open
Abstract
Inflammatory musculoskeletal diseases represent a group of chronic and disabling conditions that evolve from a complex interplay between genetic and environmental factors that cause perturbations in innate and adaptive immune responses. Understanding the pathogenesis of inflammatory musculoskeletal diseases is, to a large extent, derived from preclinical and basic research experiments. In vivo molecular imaging enables us to study molecular targets and to measure biochemical processes non-invasively and longitudinally, providing information on disease processes and potential therapeutic strategies, e.g. efficacy of novel therapeutic interventions, which is of complementary value next to ex vivo (post mortem) histopathological analysis and molecular assays. Remarkably, the large body of preclinical imaging studies in inflammatory musculoskeletal disease is in contrast with the limited reports on molecular imaging in clinical practice and clinical guidelines. Therefore, in this EANM-endorsed position paper, we performed a systematic review of the preclinical studies in inflammatory musculoskeletal diseases that involve radionuclide imaging, with a detailed description of the animal models used. From these reflections, we provide recommendations on what future studies in this field should encompass to facilitate a greater impact of radionuclide imaging techniques on the translation to clinical settings.
Collapse
Affiliation(s)
- Erik H J G Aarntzen
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Medical Imaging, Radboud University Nijmegen Medical Center, Geert Grooteplein Zuid 10, 6525 GA, Nijmegen, The Netherlands
| | - Edel Noriega-Álvarez
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine, General University Hospital of Ciudad Real, Ciudad Real, Spain
| | - Vera Artiko
- Inflammation and Infection Committee EANM, Vienna, Austria
- Center for Nuclear Medicine Clinical Center of Serbia, Faculty of Medicine, University of Belgrade, 11000, Belgrade, Serbia
| | - André H Dias
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus, Denmark
| | - Olivier Gheysens
- Inflammation and Infection Committee EANM, Vienna, Austria
- Department of Nuclear Medicine, Cliniques Universitaires Saint-Luc and Institute of Clinical and Experimental Research (IREC), Université Catholique de Louvain (UCLouvain), Brussels, Belgium
| | - Andor W J M Glaudemans
- Inflammation and Infection Committee EANM, Vienna, Austria.
- Department of Nuclear Medicine and Molecular Imaging, University Medical Center Groningen, University of Groningen Medical Imaging Center, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands.
| | - Chiara Lauri
- Inflammation and Infection Committee EANM, Vienna, Austria
- Nuclear Medicine Unit, Department of Medical-Surgical Sciences and of Translational Medicine, "Sapienza" University of Rome, 00161, Rome, Italy
| | - Giorgio Treglia
- Inflammation and Infection Committee EANM, Vienna, Austria
- Clinic of Nuclear Medicine, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
- Department of Nuclear Medicine and Molecular Imaging, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
- Faculty of Biology and Medicine, Università Della Svizzera Italiana, Lugano, Switzerland
| | - Tim van den Wyngaert
- Bone and Joint Committee EANM, Vienna, Austria
- Antwerp University Hospital Belgium, Edegem, Belgium
- Molecular Imaging Center Antwerp (MICA) - IPPON, Faculty of Medicine and Health Sciences, University of Antwerp, Universiteitsplein 1, 2610, Wilrijk, Belgium
| | - Fijs W B van Leeuwen
- Translational Molecular Imaging and Therapy Committee EANM, Vienna, Austria
- Department of Radiology, Interventional Molecular Imaging Laboratory, Leiden University Medical Center, Leiden, The Netherlands
| | - Samantha Y A Terry
- Inflammation and Infection Committee EANM, Vienna, Austria.
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, 4th Floor Lambeth Wing, St Thomas' Hospital, London, SE1 7EH, UK.
| |
Collapse
|
9
|
Tran V, Lux F, Tournier N, Jego B, Maître X, Anisorac M, Comtat C, Jan S, Selmeczi K, Evans MJ, Tillement O, Kuhnast B, Truillet C. Quantitative Tissue Pharmacokinetics and EPR Effect of AGuIX Nanoparticles: A Multimodal Imaging Study in an Orthotopic Glioblastoma Rat Model and Healthy Macaque. Adv Healthc Mater 2021; 10:e2100656. [PMID: 34212539 DOI: 10.1002/adhm.202100656] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 05/29/2021] [Indexed: 01/10/2023]
Abstract
AGuIX are emerging radiosensitizing nanoparticles (NPs) for precision radiotherapy (RT) under clinical evaluation (Phase 2). Despite being accompanied by MRI thanks to the presence of gadolinium (Gd) at its surface, more sensitive and quantifiable imaging technique should further leverage the full potential of this technology. In this study, it is shown that 89 Zr can be labeled on such NPs directly for positron emission tomography (PET) imaging with a simple and scalable method. The stability of such complexes is remarkable in vitro and in vivo. Using a glioblastoma orthotopic rat model, it is shown that injected 89 Zr-AGuIX is detectable inside the tumor for at least 1 week. Interestingly, the particles seem to efficiently infiltrate the tumor even in necrotic areas, which places great hope for the treatment of radioresistant tumor. Lastly, the first PET/MR whole-body imaging is performed in non-human primate (NHP), which further demonstrates the translational potential of these bimodal NP.
Collapse
Affiliation(s)
- Vu‐Long Tran
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - François Lux
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
- Institut Universitaire de France (IUF) Paris France
| | - Nicolas Tournier
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Benoit Jego
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Xavier Maître
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Claude Comtat
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Sébastien Jan
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | | | - Michael J. Evans
- Department of Radiology and Biomedical Imaging University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Department of Pharmaceutical Chemistry University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
- Helen Diller Family Comprehensive Cancer Center University of California San Francisco 505 Parnassus Ave San Francisco CA 94143 USA
| | - Olivier Tillement
- Institut Lumière Matière Université Claude Bernard Lyon I CNRS UMR 5306 Villeurbanne 69622 France
| | - Bertrand Kuhnast
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| | - Charles Truillet
- Laboratoire d'Imagerie Biomédicale Multimodale Paris Saclay CEA/INSERM/CNRS/Université Paris‐Saclay Orsay 91401 France
| |
Collapse
|
10
|
Wegrzyniak O, Rosestedt M, Eriksson O. Recent Progress in the Molecular Imaging of Nonalcoholic Fatty Liver Disease. Int J Mol Sci 2021; 22:7348. [PMID: 34298967 PMCID: PMC8306605 DOI: 10.3390/ijms22147348] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Revised: 06/30/2021] [Accepted: 07/06/2021] [Indexed: 12/12/2022] Open
Abstract
Pathological fibrosis of the liver is a landmark feature in chronic liver diseases, including nonalcoholic fatty liver disease (NAFLD) and nonalcoholic steatohepatitis (NASH). Diagnosis and assessment of progress or treatment efficacy today requires biopsy of the liver, which is a challenge in, e.g., longitudinal interventional studies. Molecular imaging techniques such as positron emission tomography (PET) have the potential to enable minimally invasive assessment of liver fibrosis. This review will summarize and discuss the current status of the development of innovative imaging markers for processes relevant for fibrogenesis in liver, e.g., certain immune cells, activated fibroblasts, and collagen depositions.
Collapse
Affiliation(s)
- Olivia Wegrzyniak
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; (O.W.); (M.R.)
| | - Maria Rosestedt
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; (O.W.); (M.R.)
| | - Olof Eriksson
- Science for Life Laboratory, Department of Medicinal Chemistry, Uppsala University, SE-751 83 Uppsala, Sweden; (O.W.); (M.R.)
- Antaros Medical AB, SE-431 83 Mölndal, Sweden
| |
Collapse
|
11
|
Gonzalez-Junca A, Reiners O, Borrero-Garcia LD, Beckford-Vera D, Lazar AA, Chou W, Braunstein S, VanBrocklin H, Franc BL, Barcellos-Hoff MH. Positron Emission Tomography Imaging of Functional Transforming Growth Factor β (TGFβ) Activity and Benefit of TGFβ Inhibition in Irradiated Intracranial Tumors. Int J Radiat Oncol Biol Phys 2021; 109:527-539. [PMID: 33007434 PMCID: PMC7856163 DOI: 10.1016/j.ijrobp.2020.09.043] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 09/04/2020] [Accepted: 09/21/2020] [Indexed: 12/12/2022]
Abstract
PURPOSE Transforming growth factor β (TGFβ) promotes cell survival by endorsing DNA damage repair and mediates an immunosuppressive tumor microenvironment. Thus, TGFβ activation in response to radiation therapy is potentially targetable because it opposes therapeutic control. Strategies to assess this potential in the clinic are needed. METHODS AND MATERIALS We evaluated positron emission tomography (PET) to image 89Zr -fresolimumab, a humanized TGFβ neutralizing monoclonal antibody, as a means to detect TGFβ activation in intracranial tumor models. Pathway activity of TGFβ was validated by immunodetection of phosphorylated SMAD2 and the TGFβ target, tenascin. The contribution of TGFβ to radiation response was assessed by Kaplan-Meier survival analysis of mice bearing intracranial murine tumor models GL261 and SB28 glioblastoma and brain-adapted 4T1 breast cancer (4T1-BrA) treated with TGFβ neutralizing monoclonal antibody, 1D11, and/or focal radiation (10 Gy). RESULTS 89Zr-fresolimumab PET imaging detected engineered, physiological, and radiation-induced TGFβ activation, which was confirmed by immunostaining of biological markers. GL261 glioblastoma tumors had a greater PET signal compared with similar-sized SB28 glioblastoma tumors, whereas the widespread PET signal of 4T1-BrA intracranial tumors was consistent with their highly dispersed histologic distribution. Survival of mice bearing intracranial tumors treated with 1D11 neutralizing antibody alone was similar to that of mice treated with control antibody, whereas 1D11 improved survival when given in combination with focal radiation. The extent of survival benefit of a combination of radiation and 1D11 was associated with the degree of TGFβ activity detected by PET. CONCLUSIONS This study demonstrates that 89Zr-fresolimumab PET imaging detects radiation-induced TGFβ activation in tumors. Functional imaging indicated a range of TGFβ activity in intracranial tumors, but TGFβ blockade provided survival benefit only in the context of radiation treatment. This study provides further evidence that radiation-induced TGFβ activity opposes therapeutic response to radiation.
Collapse
Affiliation(s)
- Alba Gonzalez-Junca
- Department of Radiation Oncology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Oliver Reiners
- Department of Radiation Oncology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Luis D. Borrero-Garcia
- Department of Radiation Oncology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Denis Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Ann A. Lazar
- Helen Diller Family Comprehensive Cancer Center, School of Medicine, University of California San Francisco, San Francisco, CA, USA
- Division of Oral Epidemiology, School of Dentistry, University of California San Francisco, San Francisco, CA, USA
- Division of Biostatistics, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - William Chou
- Department of Radiation Oncology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Steve Braunstein
- Department of Radiation Oncology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Henry VanBrocklin
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
| | - Benjamin L. Franc
- Department of Radiology and Biomedical Imaging, University of California San Francisco, San Francisco, CA, USA
- Current address: Department of Radiology, Stanford University, School of Medicine, Palo Alto, CA, USA
| | - Mary Helen Barcellos-Hoff
- Department of Radiation Oncology, School of Medicine, University of California San Francisco, San Francisco, CA, USA
- Helen Diller Family Comprehensive Cancer Center, School of Medicine, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
12
|
van der Krogt JMA, van Binsbergen WH, van der Laken CJ, Tas SW. Novel positron emission tomography tracers for imaging of rheumatoid arthritis. Autoimmun Rev 2021; 20:102764. [PMID: 33476822 DOI: 10.1016/j.autrev.2021.102764] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2020] [Accepted: 10/28/2020] [Indexed: 11/30/2022]
Abstract
Positron emission tomography (PET) is a nuclear imaging modality that relies on visualization of molecular targets in tissues, which is nowadays combined with a structural imaging modality such as computed tomography (CT) or Magnetic Resonance Imaging (MRI) and referred to as hybrid PET imaging. This technique allows to image specific immunological targets in rheumatoid arthritis (RA). Moreover, quantification of the PET signal enables highly sensitive monitoring of therapeutic effects on the molecular target. PET may also aid in stratification of the immuno-phenotype at baseline in order to develop personalized therapy. In this systematic review we will provide an overview of novel PET tracers, investigated in the context of RA, either pre-clinically, or clinically, that specifically visualize immune cells or stromal cells, as well as other factors and processes that contribute to pathology. The potential of these tracers in RA diagnosis, disease monitoring, and prediction of treatment outcome will be discussed. In addition, novel PET tracers established within the field of oncology that may be of use in RA will also be reviewed in order to expand the future opportunities of PET imaging in RA.
Collapse
Affiliation(s)
- Jeffrey M A van der Krogt
- Amsterdam UMC, Location AMC, Amsterdam Rheumatology & Immunology Center (ARC), University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam UMC/University of Amsterdam, Amsterdam, the Netherlands
| | - Wouter H van Binsbergen
- Amsterdam UMC, Location VUmc, Amsterdam Rheumatology and Immunology Center (ARC), VU University, Amsterdam, the Netherlands
| | - Conny J van der Laken
- Amsterdam UMC, Location VUmc, Amsterdam Rheumatology and Immunology Center (ARC), VU University, Amsterdam, the Netherlands
| | - Sander W Tas
- Amsterdam UMC, Location AMC, Amsterdam Rheumatology & Immunology Center (ARC), University of Amsterdam, Amsterdam, the Netherlands; Department of Experimental Immunology, Amsterdam UMC/University of Amsterdam, Amsterdam, the Netherlands.
| |
Collapse
|
13
|
Wang S, Li J, Hua J, Su Y, Beckford-Vera DR, Zhao W, Jayaraman M, Huynh TL, Zhao N, Wang YH, Huang Y, Qin F, Shen S, Gioeli D, Dreicer R, Sriram R, Egusa EA, Chou J, Feng FY, Aggarwal R, Evans MJ, Seo Y, Liu B, Flavell RR, He J. Molecular Imaging of Prostate Cancer Targeting CD46 Using ImmunoPET. Clin Cancer Res 2020; 27:1305-1315. [PMID: 33293372 DOI: 10.1158/1078-0432.ccr-20-3310] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 10/19/2020] [Accepted: 12/03/2020] [Indexed: 11/16/2022]
Abstract
PURPOSE We recently identified CD46 as a novel therapeutic target in prostate cancer. In this study, we developed a CD46-targeted PET radiopharmaceutical, [89Zr]DFO-YS5, and evaluated its performance for immunoPET imaging in murine prostate cancer models. EXPERIMENTAL DESIGN [89Zr]DFO-YS5 was prepared and its in vitro binding affinity for CD46 was measured. ImmunoPET imaging was conducted in male athymic nu/nu mice bearing DU145 [AR-, CD46+, prostate-specific membrane antigen-negative (PSMA-)] or 22Rv1 (AR+, CD46+, PSMA+) tumors, and in NOD/SCID gamma mice bearing patient-derived adenocarcinoma xenograft, LTL-331, and neuroendocrine prostate cancers, LTL-331R and LTL-545. RESULTS [89Zr]DFO-YS5 binds specifically to the CD46-positive human prostate cancer DU145 and 22Rv1 xenografts. In biodistribution studies, the tumor uptake of [89Zr]DFO-YS5 was 13.3 ± 3.9 and 11.2 ± 2.5 %ID/g, respectively, in DU145 and 22Rv1 xenografts, 4 days postinjection. Notably, [89Zr]DFO-YS5 demonstrated specific uptake in the PSMA- and AR-negative DU145 model. [89Zr]DFO-YS5 also showed uptake in the patient-derived LTL-331 and -331R models, with particularly high uptake in the LTL-545 neuroendocrine prostate cancer tumors (18.8 ± 5.3, 12.5 ± 1.8, and 32 ± 5.3 %ID/g in LTL-331, LTL-331R, and LTL-545, respectively, at 4 days postinjection). CONCLUSIONS [89Zr]DFO-YS5 is an excellent PET imaging agent across a panel of prostate cancer models, including in both adenocarcinoma and neuroendocrine prostate cancer, both cell line- and patient-derived xenografts, and both PSMA-positive and -negative tumors. It demonstrates potential for clinical translation as an imaging agent, theranostic platform, and companion biomarker in prostate cancer.
Collapse
Affiliation(s)
- Sinan Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Jun Li
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Department of Nuclear Medicine, Huashan Hospital, Fudan University, Shanghai, P.R. China
| | - Jun Hua
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia.,Department of Nuclear Medicine, Chongqing University Cancer Hospital, Chongqing, P.R. China
| | - Yang Su
- Department of Anesthesia, University of California, San Francisco, San Francisco, California
| | - Denis R Beckford-Vera
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Walter Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Mayuri Jayaraman
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Tony L Huynh
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Ning Zhao
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Yung-Hua Wang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Yangjie Huang
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Fujun Qin
- Department of Pathology, University of Virginia, Charlottesville, Virginia
| | - Sui Shen
- Department of Radiation Oncology, University of Alabama at Birmingham, Birmingham, Alabama
| | - Daniel Gioeli
- Department of Microbiology, Immunology, and Cancer Biology, University of Virginia, Charlottesville, Virginia.,University of Virginia Cancer Center, Charlottesville, Virginia
| | - Robert Dreicer
- University of Virginia Cancer Center, Charlottesville, Virginia.,Departments of Medicine and Urology, University of Virginia, Charlottesville, Virginia
| | - Renuka Sriram
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California
| | - Emily A Egusa
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Jonathan Chou
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Felix Y Feng
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Rahul Aggarwal
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Division of Hematology and Oncology, University of California, San Francisco, San Francisco, California
| | - Michael J Evans
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Youngho Seo
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California.,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Radiation Oncology, University of California, San Francisco, San Francisco, California
| | - Bin Liu
- Department of Anesthesia, University of California, San Francisco, San Francisco, California. .,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California
| | - Robert R Flavell
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, California. .,UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California.,Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, California
| | - Jiang He
- Department of Radiology and Medical Imaging, University of Virginia, Charlottesville, Virginia. .,University of Virginia Cancer Center, Charlottesville, Virginia
| |
Collapse
|
14
|
Giraudo C, Kainberger F, Boesen M, Trattnig S. Quantitative Imaging in Inflammatory Arthritis: Between Tradition and Innovation. Semin Musculoskelet Radiol 2020; 24:337-354. [PMID: 32992363 DOI: 10.1055/s-0040-1708823] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Radiologic imaging is crucial for diagnosing and monitoring rheumatic inflammatory diseases. Particularly the emerging approach of precision medicine has increased the interest in quantitative imaging. Extensive research has shown that ultrasound allows a quantification of direct signs such as bone erosions and synovial thickness. Dual-energy X-ray absorptiometry and high-resolution peripheral quantitative computed tomography (CT) contribute to the quantitative assessment of secondary signs such as osteoporosis or lean mass loss. Magnetic resonance imaging (MRI), using different techniques and sequences, permits in-depth evaluations. For instance, the perfusion of the inflamed synovium can be quantified by dynamic contrast-enhanced imaging or diffusion-weighted imaging, and cartilage injury can be assessed by mapping (T1ρ, T2). Furthermore, the increased metabolic activity characterizing the inflammatory response can be reliably assessed by hybrid imaging (positron emission tomography [PET]/CT, PET/MRI). Finally, advances in intelligent systems are pushing forward quantitative imaging. Complex mathematical algorithms of lesions' segmentation and advanced pattern recognition are showing promising results.
Collapse
Affiliation(s)
- Chiara Giraudo
- Department of Medicine, DIMED, Radiology Institute, University of Padova, Padova, Italy
| | - Franz Kainberger
- Division of Neuro- and Musculoskeletal Radiology, Department of Biomedical Imaging and Image-Guided Therapy, Medical University of Vienna, Vienna, Austria
| | - Mikael Boesen
- Department of Radiology, Copenhagen University Hospital Bispebjerg-Frederiksberg, Frederiksberg, Denmark
| | - Siegfried Trattnig
- Department of Biomedical Imaging and Image-Guided Therapy, High-Field MR Centre, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
15
|
Abstract
Immuno-positron emission tomography (immunoPET) is a paradigm-shifting molecular imaging modality combining the superior targeting specificity of monoclonal antibody (mAb) and the inherent sensitivity of PET technique. A variety of radionuclides and mAbs have been exploited to develop immunoPET probes, which has been driven by the development and optimization of radiochemistry and conjugation strategies. In addition, tumor-targeting vectors with a short circulation time (e.g., Nanobody) or with an enhanced binding affinity (e.g., bispecific antibody) are being used to design novel immunoPET probes. Accordingly, several immunoPET probes, such as 89Zr-Df-pertuzumab and 89Zr-atezolizumab, have been successfully translated for clinical use. By noninvasively and dynamically revealing the expression of heterogeneous tumor antigens, immunoPET imaging is gradually changing the theranostic landscape of several types of malignancies. ImmunoPET is the method of choice for imaging specific tumor markers, immune cells, immune checkpoints, and inflammatory processes. Furthermore, the integration of immunoPET imaging in antibody drug development is of substantial significance because it provides pivotal information regarding antibody targeting abilities and distribution profiles. Herein, we present the latest immunoPET imaging strategies and their preclinical and clinical applications. We also emphasize current conjugation strategies that can be leveraged to develop next-generation immunoPET probes. Lastly, we discuss practical considerations to tune the development and translation of immunoPET imaging strategies.
Collapse
Affiliation(s)
- Weijun Wei
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
| | - Zachary T Rosenkrans
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Gang Huang
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
- Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China
| | - Quan-Yong Luo
- Department of Nuclear Medicine, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai 200233, China
| | - Weibo Cai
- Departments of Radiology and Medical Physics, University of Wisconsin-Madison, 1111 Highland Avenue, Room 7137, Madison, Wisconsin 53705, United States
- Department of Pharmaceutical Sciences, University of Wisconsin-Madison, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, Madison, Wisconsin 53705, United States
| |
Collapse
|
16
|
Vermeulen K, Vandamme M, Bormans G, Cleeren F. Design and Challenges of Radiopharmaceuticals. Semin Nucl Med 2019; 49:339-356. [PMID: 31470930 DOI: 10.1053/j.semnuclmed.2019.07.001] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
This review describes general concepts with regard to radiopharmaceuticals for diagnostic or therapeutic applications that help to understand the specific challenges encountered during the design, (radio)synthesis, in vitro and in vivo evaluation and clinical translation of novel radiopharmaceuticals. The design of a radiopharmaceutical requires upfront decisions with regard to combining a suitable vector molecule with an appropriate radionuclide, considering the type and location of the molecular target, the desired application, and the time constraints imposed by the relatively short half-life of radionuclides. Well-designed in vitro and in vivo experiments allow nonclinical validation of radiotracers. Ultimately, in combination with a limited toxicology package, the radiotracer becomes a radiopharmaceutical for clinical evaluation, produced in compliance with regulatory requirements for medicines for intravenous (IV) injection.
Collapse
Affiliation(s)
- Koen Vermeulen
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Mathilde Vandamme
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| | - Guy Bormans
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium.
| | - Frederik Cleeren
- Laboratory for Radiopharmaceutical Research, Department of Pharmaceutical and Pharmacological Sciences, University of Leuven, Leuven, Belgium
| |
Collapse
|