1
|
Derlin T, Witte T. CXCR4-Targeted Imaging in Takayasu Arteritis Using 68Ga-Pentixafor PET/CT. Clin Nucl Med 2025:00003072-990000000-01727. [PMID: 40367500 DOI: 10.1097/rlu.0000000000005975] [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: 02/21/2025] [Accepted: 04/15/2025] [Indexed: 05/16/2025]
Abstract
We report a case of a 64-year-old woman with Takayasu arteritis who underwent chemokine receptor CXCR4-targeted whole-body 68Ga-Pentixafor positron emission tomography/computed tomography (PET/CT) for further evaluation of disease activity and exclusion of concurrent inflammatory diseases. 68Ga-Pentixafor PET demonstrated CXCR4-positive inflammatory cell infiltrates in the aortic wall, and active disease was further confirmed by 18F-fluorodeoxyglucose (FDG) PET/CT. Moreover, 68Ga-Pentixafor identified a CXCR4-positive pelvic bone lesion, and the patient was subsequently diagnosed with diffuse large B-cell lymphoma. CXCR4-targeted 68Ga-Pentixafor PET is useful for the detection of active inflammation in Takayasu arteritis, and to detect neoplastic disease in this cohort with increased risk of malignancy.
Collapse
Affiliation(s)
| | - Torsten Witte
- Rheumatology and Clinical Immunology, Hannover Medical School, Hanover, Germany
| |
Collapse
|
2
|
Xu T, Wan Z, Chen Y. Diffuse Uptake of 68 Ga-Pentixafor in the Breasts During the Ovulatory Phase in a Patient With SAPHO Syndrome. Clin Nucl Med 2025; 50:195-197. [PMID: 39354704 DOI: 10.1097/rlu.0000000000005459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/03/2024]
Abstract
ABSTRACT We report a case of a woman with SAPHO syndrome who exhibited increased tracer uptake in the sternal angle on a 99m Tc-MDP bone scan. This patient was enrolled in a 68 Ga-pentixafor PET/CT trial for inflammatory diseases. The PET/CT showed no abnormal tracer uptake in the sternal angle. Unexpectedly, diffuse uptake of 68 Ga-pentixafor was observed in both breasts, which might be due to hormonal stimulation because the woman underwent the 68 Ga-pentixafor PET/CT scan during the ovulatory phase.
Collapse
|
3
|
Liebich A, Bundschuh RA, Pfob CH, Kircher M, Wienand G, Raake P, Nekolla SG, Schottelius M, Higuchi T, Rieger M, Lapa C. [ 99mTc]-PentixaTec SPECT/CT for Imaging of Chemokine Receptor 4 Expression After Myocardial Infarction. Circ Cardiovasc Imaging 2024; 17:e016992. [PMID: 39534974 DOI: 10.1161/circimaging.124.016992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Accepted: 09/27/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND Accumulation of CXCR4 (C-X-C motif chemokine receptor 4)-positive immune cells after acute myocardial infarction (AMI) can be visualized by positron emission tomography. For a broader clinical application, there is a need for CXCR4-directed radiotracers labeled with isotopes that can be used with single-photon emission computed tomography (SPECT). We report on the detection of CXCR4 expression after AMI in humans using the novel tracer [99mTc]-PentixaTec. METHODS In this retrospective analysis, 9 patients with AMI after mechanical revascularization underwent myocardial inflammation imaging with [99mTc]-PentixaTec SPECT/computed tomography and rest perfusion SPECT imaging. Tracer uptake in the infarcted area, spleen, bone marrow, and blood pool were used for semiquantitative analysis and calculation of signal-to-background ratios. The extent and intensity of SPECT-derived inflammatory changes were compared with serological markers and perfusion defects. RESULTS CXCR4-directed SPECT was positive in all patients. Increased CXCR4 expression was only detected in areas with diminished perfusion corresponding to the affected vessel in coronary angiography, with a signal-to-background ratio (infarcted area-to-blood pool) of 2.36±0.74. Uptake in bone marrow and spleen showed a significant correlation with CXCR4 expression in the infarcted areas (r=0.73 and P=0.03 for spleen and r=0.81 and P=0.008 for bone marrow, respectively). The extent and intensity of SPECT-derived inflammatory changes showed no significant association with serum troponin, CK (creatine kinase), leukocyte, or CRP (C-reactive protein) levels. CONCLUSIONS This is the first report of in vivo CXCR4 imaging after AMI using a 99mTc-labeled tracer. Increased CXCR4 expression was observed locally in the infarcted region and was related to a systemic inflammatory response in the reticuloendothelial system. This proof-of-concept investigation demonstrates the general feasibility of evaluating the inflammation-related CXCR4 expression in the myocardium after AMI using conventional scintigraphy or SPECT and might, thus, broaden its worldwide application in clinical practice.
Collapse
Affiliation(s)
- Alessandro Liebich
- Nuclear Medicine, Faculty of Medicine (A.L., R.A.B., C.H.P., M.K., G.W., C.L.), University of Augsburg, Germany
| | - Ralph A Bundschuh
- Nuclear Medicine, Faculty of Medicine (A.L., R.A.B., C.H.P., M.K., G.W., C.L.), University of Augsburg, Germany
| | - Christian H Pfob
- Nuclear Medicine, Faculty of Medicine (A.L., R.A.B., C.H.P., M.K., G.W., C.L.), University of Augsburg, Germany
| | - Malte Kircher
- Nuclear Medicine, Faculty of Medicine (A.L., R.A.B., C.H.P., M.K., G.W., C.L.), University of Augsburg, Germany
| | - Georgine Wienand
- Nuclear Medicine, Faculty of Medicine (A.L., R.A.B., C.H.P., M.K., G.W., C.L.), University of Augsburg, Germany
| | - Philip Raake
- Cardiology, Faculty of Medicine (P.R., M.R.), University of Augsburg, Germany
| | - Stephan G Nekolla
- Department of Nuclear Medicine, Klinikum rechts der Isar, Technische Universität München, Germany (S.G.N.)
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and Oncology, Centre Hospitalier Universitaire Vaudois, University of Lausanne, Switzerland (M.S.)
| | - Takahiro Higuchi
- Comprehensive Heart Failure Center and Department of Nuclear Medicine, University Hospital Würzburg, Germany (T.H.)
| | - Maximilian Rieger
- Cardiology, Faculty of Medicine (P.R., M.R.), University of Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine (A.L., R.A.B., C.H.P., M.K., G.W., C.L.), University of Augsburg, Germany
| |
Collapse
|
4
|
Moghrabi S, Abdlkadir AS, Al-Hajaj N, Gnanasegaran G, Kumar R, Syed G, Bozkurt MF, Shukri S, Obeidat S, Khalaf A, Shahait M, Al-Nabhani K, Al-Ibraheem A. A New Era for PET/CT: Applications in Non-Tumorous Renal Pathologies. J Clin Med 2024; 13:4632. [PMID: 39200774 PMCID: PMC11355182 DOI: 10.3390/jcm13164632] [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: 04/25/2024] [Revised: 07/27/2024] [Accepted: 08/02/2024] [Indexed: 09/02/2024] Open
Abstract
Non-tumorous kidney diseases include a variety of conditions affecting both the structure and function of the kidneys, thereby causing a range of health-related problems. Positron emission tomography/computed tomography (PET/CT) has emerged as a potential diagnostic tool, offering a multifaceted approach to evaluating non-tumorous kidney diseases. Its clinical significance extends beyond its conventional role in cancer imaging, enabling a comprehensive assessment of renal structure and function. This review explores the diverse applications of PET/CT imaging in the evaluation of non-cancerous kidney diseases. It examines PET/CT's role in assessing acute kidney injuries, including acute pyelonephritis and other forms of nephritis, as well as chronic conditions such as immune complex-mediated glomerulonephritis and chronic kidney disease. Additionally, the review delves into PET/CT's utility in evaluating complications in renal transplant recipients, identifying renal histiocytosis and detecting renal amyloidosis. The current review aims to promote further research and technological advancements to popularize PET/CT's clinical utility in diagnosing and treating non-tumorous kidney diseases.
Collapse
Affiliation(s)
- Serin Moghrabi
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.M.); (A.S.A.); (N.A.-H.); (S.O.)
| | - Ahmed Saad Abdlkadir
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.M.); (A.S.A.); (N.A.-H.); (S.O.)
| | - Nabeela Al-Hajaj
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.M.); (A.S.A.); (N.A.-H.); (S.O.)
| | - Gopinath Gnanasegaran
- Department of Nuclear Medicine, Royal Free London NHS Foundation Trust, London NW3 2QG, UK;
| | - Rakesh Kumar
- Department of Nuclear Medicine, All India Institute of Medical Sciences, New Delhi 110608, India;
| | - Ghulam Syed
- Department of Nuclear Medicine, National Centre for Cancer Care and Research, Hamad Medical Corporation, Doha 3050, Qatar;
| | - Murat Fani Bozkurt
- Department of Nuclear Medicine, Faculty of Medicine, Hacettepe University, 06230 Ankara, Turkey;
| | - Saad Shukri
- Al-Razi Outpatient Clinic of Internal Medicine, Baghdad 10044, Iraq;
| | - Shahed Obeidat
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.M.); (A.S.A.); (N.A.-H.); (S.O.)
| | - Aysar Khalaf
- Department of Nuclear Medicine, Warith International Cancer Institute, Karbala 56001, Iraq;
| | - Mohammed Shahait
- Surgery Department, Clemenceau Medical Center, Dubai 6869, United Arab Emirates;
| | | | - Akram Al-Ibraheem
- Department of Nuclear Medicine and PET/CT, King Hussein Cancer Center (KHCC), Amman 11941, Jordan; (S.M.); (A.S.A.); (N.A.-H.); (S.O.)
- School of Medicine, University of Jordan, Amman 11942, Jordan
| |
Collapse
|
5
|
Tarchi SM, Salvatore M, Lichtenstein P, Sekar T, Capaccione K, Luk L, Shaish H, Makkar J, Desperito E, Leb J, Navot B, Goldstein J, Laifer S, Beylergil V, Ma H, Jambawalikar S, Aberle D, D'Souza B, Bentley-Hibbert S, Marin MP. Radiology of fibrosis part III: genitourinary system. J Transl Med 2024; 22:616. [PMID: 38961396 PMCID: PMC11223291 DOI: 10.1186/s12967-024-05333-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2024] [Accepted: 05/20/2024] [Indexed: 07/05/2024] Open
Abstract
Fibrosis is a pathological process involving the abnormal deposition of connective tissue, resulting from improper tissue repair in response to sustained injury caused by hypoxia, infection, or physical damage. It can impact any organ, leading to their dysfunction and eventual failure. Additionally, tissue fibrosis plays an important role in carcinogenesis and the progression of cancer.Early and accurate diagnosis of organ fibrosis, coupled with regular surveillance, is essential for timely disease-modifying interventions, ultimately reducing mortality and enhancing quality of life. While extensive research has already been carried out on the topics of aberrant wound healing and fibrogenesis, we lack a thorough understanding of how their relationship reveals itself through modern imaging techniques.This paper focuses on fibrosis of the genito-urinary system, detailing relevant imaging technologies used for its detection and exploring future directions.
Collapse
Affiliation(s)
- Sofia Maria Tarchi
- Department of Biomedical Sciences, Humanitas University, Milan, Italy.
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA.
| | - Mary Salvatore
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Philip Lichtenstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Thillai Sekar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Kathleen Capaccione
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Lyndon Luk
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hiram Shaish
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jasnit Makkar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Elise Desperito
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jay Leb
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Benjamin Navot
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Jonathan Goldstein
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sherelle Laifer
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Volkan Beylergil
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Hong Ma
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Sachin Jambawalikar
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Dwight Aberle
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Belinda D'Souza
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Stuart Bentley-Hibbert
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| | - Monica Pernia Marin
- Department of Radiology, Columbia University Irving Medical Center, 630 W 168th Street, New York, NY, 10032, USA
| |
Collapse
|
6
|
Pei J, Cheng K, Liu T, Gao M, Wang S, Xu S, Guo Y, Ma L, Li W, Wang B, Yu J, Liu J. Early, non-invasive detection of radiation-induced lung injury using PET/CT by targeting CXCR4. Eur J Nucl Med Mol Imaging 2024; 51:1109-1120. [PMID: 38030744 DOI: 10.1007/s00259-023-06517-5] [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: 08/07/2023] [Accepted: 11/06/2023] [Indexed: 12/01/2023]
Abstract
PURPOSE Radiation-induced lung injury (RILI) is a severe side effect of radiotherapy (RT) for thoracic malignancies and we currently lack established methods for the early detection of RILI. In this study, we synthesized a new tracer, [18F]AlF-NOTA-QHY-04, targeting C-X-C-chemokine-receptor-type-4 (CXCR4) and investigated its feasibility to detect RILI. METHODS An RILI rat model was constructed and scanned with [18F]AlF-NOTA-QHY-04 PET/CT and [18F]FDG PET/CT periodically after RT. Dynamic, blocking, autoradiography, and histopathological studies were performed on the day of peak uptake. Fourteen patients with radiation pneumonia, developed during or after thoracic RT, were subjected to PET scan using [18F]AlF-NOTA-QHY-04. RESULTS The yield of [18F]AlF-NOTA-QHY-04 was 28.5-43.2%, and the specific activity was 27-33 GBq/μmol. [18F]AlF-NOTA-QHY-04 was mainly excreted through the kidney. Significant increased [18F]AlF-NOTA-QHY-04 uptake in the irradiated lung compared with that in the normal lung in the RILI model was observed on day 6 post-RT and peaked on day 14 post-RT, whereas no apparent uptake of [18F]FDG was shown on days 7 and 15 post-RT. MicroCT imaging did not show pneumonia until 42 days post-RT. Significant intense [18F]AlF-NOTA-QHY-04 uptake was confirmed by autoradiography. Immunofluorescence staining demonstrated expression of CXCR4 was significantly increased in the irradiated lung tissue, which correlated with results obtained from hematoxylin-eosin and Masson's trichrome staining. In 14 patients with radiation pneumonia, maximum standardized uptake values (SUVmax) were significantly higher in the irradiated lung compared with those in the normal lung. SUVmax of patients with grade 2 RILI was significantly higher than that of patients with grade 1 RILI. CONCLUSION This study indicated that [18F]AlF-NOTA-QHY-04 PET/CT imaging can detect RILI non-invasively and earlier than [18F]FDG PET/CT in a rat model. Clinical studies verified its feasibility, suggesting the clinical potential of [18F]AlF-NOTA-QHY-04 as a PET/CT tracer for early monitoring of RILI.
Collapse
Affiliation(s)
- Jinli Pei
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Kai Cheng
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Tianxin Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Min Gao
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shijie Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shengnan Xu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Yanluan Guo
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Li Ma
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wanhu Li
- Department of PET/CT Center, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Bolin Wang
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
- Lung Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinming Yu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Jie Liu
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
7
|
Lindenberg L, Ahlman M, Lin F, Mena E, Choyke P. Advances in PET Imaging of the CXCR4 Receptor: [ 68Ga]Ga-PentixaFor. Semin Nucl Med 2024; 54:163-170. [PMID: 37923671 PMCID: PMC10792730 DOI: 10.1053/j.semnuclmed.2023.09.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023]
Abstract
[68Ga]Ga-PentixaFor, a PET agent targeting CXCR4 is emerging as a versatile radiotracer with promising applications in oncology, cardiology and inflammatory disease. Preclinical work in various cancer cell lines have demonstrated high specificity and selectivity. In human investigations of several tumors, the most promising applications may be in multiple myeloma, certain lymphomas and myeloproliferative neoplasms. In the nononcologic setting, [68Ga]Ga-PentixaFor could greatly improve detection for primary aldosteronism and other endocrine abnormalities. Similarly, atherosclerotic disease and other inflammatory conditions could also benefit from enhanced identification by CXCR4 targeting. Rapidly cleared from the body with a favorable imaging and radiation dosimetry profile that has been already studied in over 1000 patients, [68Ga]Ga-PentixaFor is a worthy agent for further clinical exploration with potential for theranostic applications in hematologic malignancies.
Collapse
Affiliation(s)
- Liza Lindenberg
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD; Uniformed Services University of the Health Sciences, Bethesda, MD.
| | - Mark Ahlman
- Department of Radiology and Imaging, Medical College of Georgia, Augusta, GA
| | - Frank Lin
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Esther Mena
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Peter Choyke
- Molecular Imaging Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| |
Collapse
|
8
|
Konrad M, Rinscheid A, Wienand G, Nittbaur B, Wester HJ, Janzen T, Lapa C, Pfob CH, Schottelius M. [ 99mTc]Tc-PentixaTec: development, extensive pre-clinical evaluation, and first human experience. Eur J Nucl Med Mol Imaging 2023; 50:3937-3948. [PMID: 37597009 PMCID: PMC10611619 DOI: 10.1007/s00259-023-06395-x] [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: 06/02/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023]
Abstract
PURPOSE The clinical success non-invasive imaging of CXCR4 expression using [68 Ga]Ga-PentixaFor-PET warrants an expansion of the targeting concept towards conventional scintigraphy/SPECT with their lower cost and general availability. To this aim, we developed and comparatively evaluated a series of 99mTc-labeled cyclic pentapeptides based on the PentixaFor scaffold. METHODS Six mas3-conjugated CPCR4 analogs with different 4-aminobenzoic acid (Abz)-D-Ala-D-Arg-aa3 linkers (L1-L6) as well as the corresponding HYNIC- and N4-analogs of L6-CPCR4 were synthesized via standard SPPS. Competitive binding studies (IC50 and IC50inv) were carried out using Jurkat T cell lymphoma cells and [125I]FC-131 as radioligand. Internalization kinetics were investigated using hCXCR4-overexpressing Chem-1 cells. Biodistribution studies and small animal SPECT/CT imaging (1 h p.i.) were carried out using Jurkat xenograft bearing CB17/SCID mice. Based on the preclinical results, [99mTc]Tc-N4-L6-CPCR4 ([99mTc]Tc-PentixaTec) was selected for an early translation to the human setting. Five patients with hematologic malignancies underwent [99mTc]Tc-N4-L6-CPCR4 SPECT/planar imaging with individual dosimetry. RESULTS Of the six mas3-conjugated peptides, mas3-L6-CPCR4 (mas3-dap-r-a-Abz-CPCR4) showed the highest CXCR4 affinity (IC50 = 5.0 ± 1.3 nM). Conjugation with N4 (N4-L6-CPCR4) further improved hCXCR4 affinity to 0.6 ± 0.1 nM. [99mTc]Tc-N4-L6-CPCR4 also showed the most efficient internalization (97% of total cellular activity at 2 h) and the highest tumor accumulation (8.6 ± 1.3% iD/g, 1 h p.i.) of the compounds investigated. Therefore, [99mTc]Tc-N4-L6-CPCR4 (termed [99mTc]Tc-PentixaTec) was selected for first-in-human application. [99mTc]Tc-PentixaTec was well tolerated, exhibits a favorable biodistribution and dosimetry profile (2.1-3.4 mSv per 500 MBq) and excellent tumor/background ratios in SPECT and planar imaging. CONCLUSION The successive optimization of the amino acid composition of the linker structure and the N-terminal 99mTc-labeling strategies (mas3 vs HYNIC vs N4) has provided [99mTc]Tc-PentixaTec as a novel, highly promising CXCR4-targeted SPECT agent for clinical application. With its excellent CXCR4 affinity, efficient internalization, high uptake in CXCR4-expressing tissues, suitable clearance/biodistribution characteristics, and favorable human dosimetry, it holds great potential for further clinical use.
Collapse
Affiliation(s)
- Matthias Konrad
- Chair for Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München, 85748, Garching, Germany
| | - Andreas Rinscheid
- Medical Physics and Radiation Protection, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Georgine Wienand
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Bernd Nittbaur
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Hans-Jürgen Wester
- Chair for Pharmaceutical Radiochemistry, Faculties of Chemistry and Medicine, Technische Universität München, 85748, Garching, Germany
| | - Tilman Janzen
- Medical Physics and Radiation Protection, University Hospital Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Constantin Lapa
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany
| | - Christian Helmut Pfob
- Nuclear Medicine, Faculty of Medicine, University of Augsburg, Stenglinstrasse 2, 86156, Augsburg, Germany.
| | - Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Rue du Bugnon 25A, Agora, CH-1011, Lausanne, Switzerland.
- AGORA, Pôle de Recherche Sur Le Cancer, 1011, Lausanne, Switzerland.
- SCCL Swiss Cancer Center Leman, 1011, Lausanne, Switzerland.
| |
Collapse
|
9
|
Higuchi T, Hartrampf PE, Buck AK, Pomper MG, Rowe SP, Serfling SE, Werner RA. Role of Functional SPECT and PET in Renal Emergencies. Semin Nucl Med 2023; 53:786-796. [PMID: 37236903 DOI: 10.1053/j.semnuclmed.2023.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 04/14/2023] [Indexed: 05/28/2023]
Abstract
Renal scintigraphy is a centerpiece of nuclear medicine and is also commonly applied for (peri-)acute care. In this regard, referrals by the treating physician include: I.) acute obstructions caused by gradual and infiltrative tumor growth or renal off-target effects under anti-tumor treatment, II.) functional issues in infants, for example, structural abnormalities such as duplex kidneys or uroliths in adults, which can also trigger III.) Infections of renal parenchyma. Renal radionuclide imaging is also requested due to IV.) acute trauma to the abdomen, for example, to assess renal scarring or upon further follow-up after reconstructive surgery. We will discuss clinical applications of (peri-)acute renal scintigraphy, along with future prospects on the use of more advanced nuclear imaging techniques such as renal positron emission tomography.
Collapse
Affiliation(s)
- Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany; Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan.
| | - Philipp E Hartrampf
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany
| | - Martin G Pomper
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | - Steven P Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| | | | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Würzburg, Germany; Comprehensive Heart Failure Center, University Hospital Würzburg, Würzburg, Germany; Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD
| |
Collapse
|
10
|
Nair A, Pandit N, Kavadichanda C. Role of [68Ga]-pentixafor positron emission tomography/computed tomography imaging in assessing disease activity in patients with lupus nephritis: A pilot study. Lupus 2023; 32:1267-1275. [PMID: 37691452 DOI: 10.1177/09612033231201625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/12/2023]
Abstract
OBJECTIVE The primary objective of our study was to evaluate renal uptake of 68Ga-pentixafor in patients with lupus nephritis. Eighteen patients who satisfied the inclusion criteria were included in our study. METHODS The study participants were patients with histopathologically confirmed lupus nephritis who were referred to our department for 68Ga-pentixafor PET/CT scan. We studied the renal images in these patients for uptake patterns based on purely visual as well as semi-quantitative parameters. The visual parameters included uptake relative to the spleen and liver. Semi-quantitative analysis involved the uptake as given by SUVmax. These parameters were correlated with the patients' biochemical as well as histological parameters. Kendall's tau-b test was used to look for an association between renal uptake by visual assessment and histopathological findings. Mean SUVmax values were compared by using the Mann-Whitney U test and a p value < .05 was considered to be statistically significant. RESULTS No significant association between the mean renal SUVmax of the bilateral kidneys in pentixafor PET and histopathological class was observed. We did not observe any heterogeneity in uptake patterns that could be attributed to the disease process in our patients. CONCLUSION 68Ga-pentixafor PET is not a suitable imaging modality for assessment of disease activity in lupus nephritis patients. There is a void in non-invasive testing for patients with this chronic and often disabling condition which calls for further research in this area.
Collapse
Affiliation(s)
- Ahalya Nair
- Department of Nuclear Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Nandini Pandit
- Department of Nuclear Medicine, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| | - Chengappa Kavadichanda
- Department of Clinical Immunology, Jawaharlal Institute of Postgraduate Medical Education and Research (JIPMER), Puducherry, India
| |
Collapse
|
11
|
Predicting Microenvironment in CXCR4- and FAP-Positive Solid Tumors-A Pan-Cancer Machine Learning Workflow for Theranostic Target Structures. Cancers (Basel) 2023; 15:cancers15020392. [PMID: 36672341 PMCID: PMC9856808 DOI: 10.3390/cancers15020392] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 01/04/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
(1) Background: C-X-C Motif Chemokine Receptor 4 (CXCR4) and Fibroblast Activation Protein Alpha (FAP) are promising theranostic targets. However, it is unclear whether CXCR4 and FAP positivity mark distinct microenvironments, especially in solid tumors. (2) Methods: Using Random Forest (RF) analysis, we searched for entity-independent mRNA and microRNA signatures related to CXCR4 and FAP overexpression in our pan-cancer cohort from The Cancer Genome Atlas (TCGA) database-representing n = 9242 specimens from 29 tumor entities. CXCR4- and FAP-positive samples were assessed via StringDB cluster analysis, EnrichR, Metascape, and Gene Set Enrichment Analysis (GSEA). Findings were validated via correlation analyses in n = 1541 tumor samples. TIMER2.0 analyzed the association of CXCR4 / FAP expression and infiltration levels of immune-related cells. (3) Results: We identified entity-independent CXCR4 and FAP gene signatures representative for the majority of solid cancers. While CXCR4 positivity marked an immune-related microenvironment, FAP overexpression highlighted an angiogenesis-associated niche. TIMER2.0 analysis confirmed characteristic infiltration levels of CD8+ cells for CXCR4-positive tumors and endothelial cells for FAP-positive tumors. (4) Conclusions: CXCR4- and FAP-directed PET imaging could provide a non-invasive decision aid for entity-agnostic treatment of microenvironment in solid malignancies. Moreover, this machine learning workflow can easily be transferred towards other theranostic targets.
Collapse
|
12
|
Werner RA, Pomper MG, Buck AK, Rowe SP, Higuchi T. SPECT and PET Radiotracers in Renal Imaging. Semin Nucl Med 2022; 52:406-418. [DOI: 10.1053/j.semnuclmed.2021.12.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 12/06/2021] [Indexed: 12/24/2022]
|
13
|
Schottelius M, Herrmann K, Lapa C. In Vivo Targeting of CXCR4-New Horizons. Cancers (Basel) 2021; 13:5920. [PMID: 34885030 PMCID: PMC8656854 DOI: 10.3390/cancers13235920] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 01/23/2023] Open
Abstract
Given its pre-eminent role in the context of tumor cell growth as well as metastasis, the C-X-C motif chemokine receptor 4 (CXCR4) has attracted a lot of interest in the field of nuclear oncology, and clinical evidence on the high potential of CXCR4-targeted theranostics is constantly accumulating. Additionally, since CXCR4 also represents a key player in the orchestration of inflammatory responses to inflammatory stimuli, based on its expression on a variety of pro- and anti-inflammatory immune cells (e.g., macrophages and T-cells), CXCR4-targeted inflammation imaging has recently gained considerable attention. Therefore, after briefly summarizing the current clinical status quo of CXCR4-targeted theranostics in cancer, this review primarily focuses on imaging of a broad spectrum of inflammatory diseases via the quantification of tissue infiltration with CXCR4-expressing immune cells. An up-to-date overview of the ongoing preclinical and clinical efforts to visualize inflammation and its resolution over time is provided, and the predictive value of the CXCR4-associated imaging signal for disease outcome is discussed. Since the sensitivity and specificity of CXCR4-targeted immune cell imaging greatly relies on the availability of suitable, tailored imaging probes, recent developments in the field of CXCR4-targeted imaging agents for various applications are also addressed.
Collapse
Affiliation(s)
- Margret Schottelius
- Translational Radiopharmaceutical Sciences, Department of Nuclear Medicine and of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV), University of Lausanne (UNIL), 1011 Lausanne, Switzerland
| | - Ken Herrmann
- Department of Nuclear Medicine, German Cancer Consortium (DKTK)-University Hospital Essen, University of Duisburg-Essen, 45147 Essen, Germany;
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, 86156 Augsburg, Germany
| |
Collapse
|
14
|
The utilization of positron emission tomography in the evaluation of renal health and disease. Clin Transl Imaging 2021. [DOI: 10.1007/s40336-021-00469-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Abstract
Purpose
Positron emission tomography (PET) is a nuclear imaging technique that uses radiotracers to visualize metabolic processes of interest across different organs, to diagnose and manage diseases, and monitor therapeutic response. This systematic review aimed to characterize the value of PET for the assessment of renal metabolism and function in subjects with non-oncological metabolic disorders.
Methods
This review was conducted and reported in accordance with the PRISMA statement. Research articles reporting “kidney” or “renal” metabolism evaluated with PET imaging between 1980 and 2021 were systematically searched in Medline/PubMed, Science Direct, and the Cochrane Library. Search results were exported and stored in RefWorks, the duplicates were removed, and eligible studies were identified, evaluated, and summarized.
Results
Thirty reports met the inclusion criteria. The majority of the studies were prospective (73.33%, n = 22) in nature. The most utilized PET radiotracers were 15O-labeled radio water (H215O, n = 14) and 18F-fluorodeoxyglucose (18F-FDG, n = 8). Other radiotracers used in at least one study were 14(R,S)-(18)F-fluoro-6-thia-heptadecanoic acid (18F-FTHA), 18F-Sodium Fluoride (18F-NaF), 11C-acetate, 68-Gallium (68Ga), 13N-ammonia (13N-NH3), Rubidium-82 (82Rb), radiolabeled cationic ferritin (RadioCF), 11C‐para-aminobenzoic acid (11C-PABA), Gallium-68 pentixafor (68Ga-Pentixafor), 2-deoxy-2-F-fluoro-d-sorbitol (F-FDS) and 55Co-ethylene diamine tetra acetic acid (55Co-EDTA).
Conclusion
PET imaging provides an effective modality for evaluating a range of metabolic functions including glucose and fatty acid uptake, oxygen consumption and renal perfusion. Multiple positron emitting radiolabeled racers can be used for renal imaging in clinical settings. PET imaging thus holds the potential to improve the diagnosis of renal disorders, and to monitor disease progression and treatment response.
Collapse
|
15
|
Klinkhammer BM, Lammers T, Mottaghy FM, Kiessling F, Floege J, Boor P. Non-invasive molecular imaging of kidney diseases. Nat Rev Nephrol 2021; 17:688-703. [PMID: 34188207 PMCID: PMC7612034 DOI: 10.1038/s41581-021-00440-4] [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] [Accepted: 05/10/2021] [Indexed: 02/05/2023]
Abstract
In nephrology, differential diagnosis or assessment of disease activity largely relies on the analysis of glomerular filtration rate, urinary sediment, proteinuria and tissue obtained through invasive kidney biopsies. However, currently available non-invasive functional parameters, and most serum and urine biomarkers, cannot capture intrarenal molecular disease processes specifically. Moreover, although histopathological analyses of kidney biopsy samples enable the visualization of pathological morphological and molecular alterations, they only provide information about a small part of the kidney and do not allow longitudinal monitoring. These limitations not only hinder understanding of the dynamics of specific disease processes in the kidney, but also limit the targeting of treatments to active phases of disease and the development of novel targeted therapies. Molecular imaging enables non-invasive and quantitative assessment of physiological or pathological processes by combining imaging technologies with specific molecular probes. Here, we discuss current preclinical and clinical molecular imaging approaches in nephrology. Non-invasive visualization of the kidneys through molecular imaging can be used to detect and longitudinally monitor disease activity and can therefore provide companion diagnostics to guide clinical trials, as well as the safe and effective use of drugs.
Collapse
Affiliation(s)
| | - Twan Lammers
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
- Department of Pharmaceutics, Utrecht University, Utrecht, Netherlands
- Department of Targeted Therapeutics, University of Twente, Enschede, Netherlands
| | - Felix M Mottaghy
- Department of Nuclear Medicine, University Hospital RWTH, Aachen, Germany
- Department of Radiology and Nuclear Medicine, Maastricht University Medical Center, Maastricht, Netherlands
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University Hospital, Aachen, Germany
- Fraunhofer Institute for Digital Medicine MEVIS, Bremen, Germany
| | - Jürgen Floege
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Hospital, Aachen, Germany.
- Department of Nephrology and Immunology, RWTH Aachen University Hospital, Aachen, Germany.
- Electron Microscopy Facility, RWTH Aachen University Hospital, Aachen, Germany.
| |
Collapse
|
16
|
Werner RA, Hess A, Koenig T, Diekmann J, Derlin T, Melk A, Thackeray JT, Bauersachs J, Bengel FM. Molecular imaging of inflammation crosstalk along the cardio-renal axis following acute myocardial infarction. Theranostics 2021; 11:7984-7994. [PMID: 34335975 PMCID: PMC8315063 DOI: 10.7150/thno.61423] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2021] [Accepted: 05/05/2021] [Indexed: 01/10/2023] Open
Abstract
Rationale: Acute myocardial infarction (MI) triggers a systemic inflammatory response including crosstalk along the heart-kidney axis. We employed radionuclide-based inflammation-targeted whole-body molecular imaging to identify potential cardio-renal crosstalk after MI in a translational setup. Methods: Serial whole-body positron emission tomography (PET) with the specific CXCR4 ligand 68Ga-Pentixafor was performed after MI in mice. Tracer retention in kidneys and heart was compared to hematopoietic organs to evaluate systemic inflammation, validated by ex vivo analysis and correlated with progressive contractile dysfunction. Additionally, 96 patients underwent 68Ga-Pentixafor PET within the first week after MI, for systems-based image analysis and to determine prognostic value for adverse renal outcome. Results: In mice, transient myocardial CXCR4 upregulation occurred early after MI. Cardiac and renal PET signal directly correlated over the time course (r = 0.62, p < 0.0001), suggesting an inflammatory link between organs. Ex-vivo autoradiography (r = 0.9, p < 0.01) and CD68 immunostaining indicated signal localization to inflammatory cell content. Renal signal at 7d was inversely proportional to left ventricular ejection fraction at 6 weeks after MI (r = -0.79, p < 0.01). In patients, renal CXCR4 signal also correlated with signal from infarct (r = 0.25, p < 0.05) and remote myocardium (r = 0.39, p < 0.0001). Glomerular filtration rate (GFR) was available in 48/96 (50%) during follow-up. Worsening of renal function (GFR loss >5 mL/min/1.73m2), occurred a mean 80.5 days after MI in 16/48 (33.3%). Kaplan-Meier analysis revealed adverse renal outcome for patients with elevated remote myocardial CXCR4 signal (p < 0.05). Multivariate Cox analysis confirmed an independent predictive value (relative to baseline GFR, LVEF, infarct size; HR, 5.27). Conclusion: Systems-based CXCR4-targeted molecular imaging identifies inflammatory crosstalk along the cardio-renal axis early after MI.
Collapse
Affiliation(s)
- Rudolf A. Werner
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Annika Hess
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Tobias Koenig
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Johanna Diekmann
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Anette Melk
- Department of Kidney, Liver and Metabolic Diseases, Children's Hospital, Hannover Medical School, Hannover, Germany
| | - James T. Thackeray
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Johann Bauersachs
- Department of Cardiology and Angiology, Hannover Medical School, Hannover, Germany
| | - Frank M. Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| |
Collapse
|
17
|
Caroli A, Remuzzi A, Lerman LO. Basic principles and new advances in kidney imaging. Kidney Int 2021; 100:1001-1011. [PMID: 33984338 DOI: 10.1016/j.kint.2021.04.032] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 04/21/2021] [Accepted: 04/22/2021] [Indexed: 12/12/2022]
Abstract
Over the past few years, clinical renal imaging has seen great advances, allowing assessments of kidney structure and morphology, perfusion, function and metabolism, and oxygenation, as well as microstructure and the interstitium. Medical imaging is becoming increasingly important in the evaluation of kidney physiology and pathophysiology, showing promise in management of patients with renal disease, in particular with regard to diagnosis, classification, and prediction of disease development and progression, monitoring response to therapy, detection of drug toxicity, and patient selection for clinical trials. A variety of imaging modalities, ranging from routine to advanced tools, are currently available to probe the kidney both spatially and temporally, particularly ultrasonography, computed tomography, positron emission tomography, renal scintigraphy, and multiparametric magnetic resonance imaging. Given that the range is broad and varied, kidney imaging techniques should be chosen based on the clinical question and the specific underlying pathologic mechanism, taking into account contraindications and possible adverse effects. Integration of various modalities providing complementary information will likely provide the greatest insight into renal pathophysiology. This review aims to highlight major recent advances in key tools that are currently available or potentially relevant for clinical kidney imaging, with a focus on non-oncological applications. The review also outlines the context of use, limitations, and advantages of various techniques, and highlights gaps to be filled with future development and clinical adoption.
Collapse
Affiliation(s)
- Anna Caroli
- Bioengineering Department, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Bergamo, Italy.
| | - Andrea Remuzzi
- Department of Management, Information and Production Engineering, University of Bergamo, Dalmine (Bergamo), Italy
| | - Lilach O Lerman
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
18
|
Toyama Y, Werner RA, Ruiz-Bedoya CA, Ordonez AA, Takase K, Lapa C, Jain SK, Pomper MG, Rowe SP, Higuchi T. Current and future perspectives on functional molecular imaging in nephro-urology: theranostics on the horizon. Theranostics 2021; 11:6105-6119. [PMID: 33897902 PMCID: PMC8058716 DOI: 10.7150/thno.58682] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 03/21/2021] [Indexed: 01/01/2023] Open
Abstract
In recent years, a paradigm shift from single-photon-emitting radionuclide radiotracers toward positron-emission tomography (PET) radiotracers has occurred in nuclear oncology. Although PET-based molecular imaging of the kidneys is still in its infancy, such a trend has emerged in the field of functional renal radionuclide imaging. Potentially allowing for precise and thorough evaluation of renal radiotracer urodynamics, PET radionuclide imaging has numerous advantages including precise anatomical co-registration with CT images and dynamic three-dimensional imaging capability. In addition, relative to scintigraphic approaches, PET can allow for significantly reduced scan time enabling high-throughput in a busy PET practice and further reduces radiation exposure, which may have a clinical impact in pediatric populations. In recent years, multiple renal PET radiotracers labeled with 11C, 68Ga, and 18F have been utilized in clinical studies. Beyond providing a precise non-invasive read-out of renal function, such radiotracers may also be used to assess renal inflammation. This manuscript will provide an overview of renal molecular PET imaging and will highlight the transformation of conventional scintigraphy of the kidneys toward novel, high-resolution PET imaging for assessing renal function. In addition, future applications will be introduced, e.g. by transferring the concept of molecular image-guided diagnostics and therapy (theranostics) to the field of nephrology.
Collapse
Affiliation(s)
- Yoshitaka Toyama
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Department of Diagnostic Radiology, Tohoku University, Sendai, Japan
| | - Rudolf A. Werner
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, Wuerzburg Germany
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Camilo A. Ruiz-Bedoya
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Alvaro A. Ordonez
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Kei Takase
- Department of Diagnostic Radiology, Tohoku University, Sendai, Japan
| | - Constantin Lapa
- Nuclear Medicine, Medical Faculty, University of Augsburg, Augsburg, Germany
| | - Sanjay K. Jain
- Center for Infection and Inflammation Imaging Research, Johns Hopkins University School of Medicine, Baltimore, Maryland
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Martin G. Pomper
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Steven P. Rowe
- Division of Nuclear Medicine and Molecular Imaging, The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- The James Buchanan Brady Urological Institute and Department of Urology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Takahiro Higuchi
- Department of Nuclear Medicine, University Hospital Wuerzburg, Wuerzburg, Germany
- Comprehensive Heart Failure Center, University Hospital Wuerzburg, Wuerzburg Germany
- Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama, Japan
| |
Collapse
|
19
|
Imaging Inflammation with Positron Emission Tomography. Biomedicines 2021; 9:biomedicines9020212. [PMID: 33669804 PMCID: PMC7922638 DOI: 10.3390/biomedicines9020212] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2021] [Revised: 01/28/2021] [Accepted: 02/12/2021] [Indexed: 12/19/2022] Open
Abstract
The impact of inflammation on the outcome of many medical conditions such as cardiovascular diseases, neurological disorders, infections, cancer, and autoimmune diseases has been widely acknowledged. However, in contrast to neurological, oncologic, and cardiovascular disorders, imaging plays a minor role in research and management of inflammation. Imaging can provide insights into individual and temporospatial biology and grade of inflammation which can be of diagnostic, therapeutic, and prognostic value. There is therefore an urgent need to evaluate and understand current approaches and potential applications for imaging of inflammation. This review discusses radiotracers for positron emission tomography (PET) that have been used to image inflammation in cardiovascular diseases and other inflammatory conditions with a special emphasis on radiotracers that have already been successfully applied in clinical settings.
Collapse
|
20
|
Abstract
Renal transplantation has become the best treatment for the patients with chronic renal insufficiency. The surgical procedures, immunosuppressive regiments and patient follow-up have evolved especially in the last 10 years. However, the diagnosis for renal transplantation dysfunction remained the same in these years. Serum creatinine levels and estimated glomerular filtration rate calculated by serum creatinine based equations are used in routine patient follow-up. Pelvic ultrasonography and color Doppler ultrasonography are used as a first-line imaging method. Assessment of allograft functions both qualitatively and quantitatively are possible using nuclear medicine procedures. Surgical complications, acute tubular necrosis, subacute and/or acute rejection, infections, toxicity due to immunosuppressive medications, complications relating the collecting system, chronic rejection are the main causes for renal function impairment. The imaging procedures can diagnose the worsening of renal transplant function; however, they still lack the ability to differentiate types of rejection as histopathology or differentiate rejection from other causes of allograft dysfunction. The transplant biopsy gives detailed diagnosis for allograft dysfunction, guide the treatment and therefore it is the preferred diagnostic choice in recent years. On recent years, literature on radionuclide imaging is focused on perfusion analysis for the early diagnosis of renal transplant dysfunction and prognostic use of perfusion parameters, and then this article will focus on these studies and their outcome.
Collapse
Affiliation(s)
- Bilge Volkan-Salanci
- Assoc Prof. Dept. of Nuclear Medicine, Hacettepe University, Medical School, Ankara, Turkey
| | - Belkis Erbas
- Prof. Dept. of Nuclear Medicine, Hacettepe University, Medical School, Ankara, Turkey.
| |
Collapse
|
21
|
Pan Q, Luo Y, Cao X, Li J. Increased Uptake of 68Ga-Pentixafor in Thymic Rebound After Chemotherapy in a Patient With Multiple Myeloma. Clin Nucl Med 2021; 46:90-92. [PMID: 33181744 DOI: 10.1097/rlu.0000000000003375] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
A 38-year-old man with newly diagnosed multiple myeloma underwent Ga-pentixafor PET/CT for baseline assessment. Ga-pentixafor PET/CT demonstrated multiple focal bone and bone marrow lesions with intense radioactivity. After 9 cycles of chemotherapy against multiple myeloma, the patient experienced serologically complete remission of myeloma. Consistently, the follow-up Ga-pentixafor PET/CT for PET response showed significantly reduced uptake in the previously chemokine receptor 4-positive bone lesions. However, intense and homogeneous uptake of Ga-pentixafor was noted in the thymus with a smooth contour and slightly increased density, which is possibly caused by thymic hyperplasia after chemotherapy.
Collapse
Affiliation(s)
| | | | - Xinxin Cao
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| | - Jian Li
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, China
| |
Collapse
|
22
|
Derlin T, Jaeger B, Jonigk D, Apel RM, Freise J, Shin HO, Weiberg D, Warnecke G, Ross TL, Wester HJ, Seeliger B, Welte T, Bengel FM, Prasse A. Clinical Molecular Imaging of Pulmonary CXCR4 Expression to Predict Outcome of Pirfenidone Treatment in Idiopathic Pulmonary Fibrosis. Chest 2020; 159:1094-1106. [PMID: 32822674 DOI: 10.1016/j.chest.2020.08.2043] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2020] [Revised: 08/03/2020] [Accepted: 08/10/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Idiopathic pulmonary fibrosis (IPF) is a progressive disease for which two antifibrotic drugs recently were approved. However, an unmet need exists to predict responses to antifibrotic treatment, such as pirfenidone. Recent data suggest that upregulated expression of CXCR4 is indicative of outcomes in IPF. RESEARCH QUESTION Can quantitative, molecular imaging of pulmonary CXCR4 expression as a biomarker for disease activity predict response to the targeted treatment pirfenidone and prognosis in patients with IPF? STUDY DESIGN AND METHODS CXCR4 expression was analyzed by immunohistochemistry examination of lung tissues and reverse-transcriptase polymerase chain reaction analysis of BAL. PET-CT scanning with the specific CXCR4 ligand 68Ga-pentixafor was performed in 28 IPF patients and compared with baseline clinical characteristics. In 16 patients, a follow-up scan was obtained 6 to 12 weeks after initiation of treatment with pirfenidone. Patients were followed up in our outpatient clinic for ≥ 12 months. RESULTS Immunohistochemistry analysis showed high CXCR4 staining of epithelial cells and macrophages in areas with vast fibrotic remodeling. Targeted PET scanning revealed CXCR4 upregulation in fibrotic areas of the lungs, particularly in zones with subpleural honeycombing. Baseline CXCR4 signal demonstrated a significant correlation with Gender Age Physiology stage (r = 0.44; P = .02) and with high-resolution CT scan score (r = 0.38; P = .04). Early changes in CXCR4 signal after initiation of pirfenidone treatment correlated with the long-term course of FVC after 12 months (r = -0.75; P = .0008). Moreover, patients with a high pulmonary CXCR4 signal on follow-up PET scan after 6 weeks into treatment demonstrated a statistically significant worse outcome at 12 months (P = .002). In multiple regression analysis, pulmonary CXCR4 signal on follow-up PET scan emerged as the only independent predictor of long-term outcome (P = .0226). INTERPRETATION CXCR4-targeted PET imaging identified disease activity and predicted outcome of IPF patients treated with pirfenidone. It may serve as a future biomarker for personalized guidance of antifibrotic treatment.
Collapse
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Benedikt Jaeger
- Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany
| | - Danny Jonigk
- Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Rosa M Apel
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Julia Freise
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Hoen-Oh Shin
- Institute of Radiology, Hannover Medical School, Hannover, Germany
| | - Desiree Weiberg
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Gregor Warnecke
- Department of Heart, Thoracic, Transplantation and Vascular Surgery, Hannover Medical School, Hannover, Germany
| | - Tobias L Ross
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Hans-Jürgen Wester
- Institute of Radiopharmaceutical Chemistry, Technical University Munich, Garching, Germany
| | - Benjamin Seeliger
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Tobias Welte
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; DZL-BREATH, Hannover, Germany
| | - Frank M Bengel
- Department of Nuclear Medicine, Hannover Medical School, Hannover, Germany
| | - Antje Prasse
- Department of Pulmonology, Hannover Medical School, Hannover, Germany; Fraunhofer Institute for Toxicology and Experimental Medicine, Hannover, Germany; DZL-BREATH, Hannover, Germany.
| |
Collapse
|
23
|
Osl T, Schmidt A, Schwaiger M, Schottelius M, Wester HJ. A new class of PentixaFor- and PentixaTher-based theranostic agents with enhanced CXCR4-targeting efficiency. Am J Cancer Res 2020; 10:8264-8280. [PMID: 32724470 PMCID: PMC7381729 DOI: 10.7150/thno.45537] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2020] [Accepted: 06/05/2020] [Indexed: 02/07/2023] Open
Abstract
Non-invasive PET imaging of CXCR4 expression in cancer and inflammation as well as CXCR4-targeted radioligand therapy (RLT) have recently found their way into clinical research by the development of the theranostic agents [68Ga]PentixaFor (cyclo(D-Tyr1-D-[NMe]Orn2(AMBS-[68Ga]DOTA)-Arg3-Nal4-Gly5) = [68Ga]DOTA-AMBS-CPCR4) and [177Lu/90Y]PentixaTher (cyclo(D-3-iodo-Tyr1-D-[NMe]Orn2(AMBS-[177Lu/90Y]DOTA)-Arg3-Nal4-Gly5) = [177Lu/90Y]DOTA-AMBS-iodoCPCR4). Although convincing clinical results have already been obtained with both agents, this study was designed to further investigate the required structural elements for improved ligand-receptor interaction for both peptide cores (CPCR4 and iodoCPCR4). To this aim, a series of DOTA-conjugated CPCR4- and iodoCPCR4-based ligands with new linker structures, replacing the AMBA-linker in PentixaFor and PentixaTher, were synthesized and evaluated. Methods: The in vitro investigation of the novel compounds alongside with the reference peptides PentixaFor and PentixaTher encompassed the determination of hCXCR4 and mCXCR4 affinity (IC50) of the respective natGa-, natLu-, natY- and natBi-complexes in Jurkat and Eμ-myc 1080 cells using [125I]FC-131 and [125I]CPCR4.3 as radioligands, respectively, as well as the evaluation of the internalization and externalization kinetics of selected 68Ga- and 177Lu-labeled compounds in hCXCR4-transfected Chem-1 cells. Comparative small animal PET imaging studies (1h p.i.) as well as in vivo biodistribution studies (1, 6 and 48h p.i.) were performed in Daudi (human B cell lymphoma) xenograft bearing CB17 SCID mice. Results: Based on the affinity data and cellular uptake studies, [68Ga/177Lu]DOTA-r-a-ABA-CPCR4 and [68Ga/177Lu]DOTA-r-a-ABA-iodoCPCR4 (with r-a-ABA = D-Arg-D-Ala-4-aminobenzoyl-) were selected for further evaluation. Both analogs show app. 10-fold enhanced hCXCR4 affinity compared to the respective references [68Ga]PentixaFor and [177Lu]PentixaTher, four times higher cellular uptake in hCXCR4 expressing cells and improved cellular retention. Unfortunately, the improved in vitro binding and uptake characteristics of [68Ga]DOTA-r-a-ABA-CPCR4 and -iodoCPCR4 could not be recapitulated in initial PET imaging studies; both compounds showed similar uptake in the Daudi xenografts as [68Ga]PentixaFor, alongside with higher background accumulation, especially in the kidneys. However, the subsequent biodistribution studies performed for the corresponding 177Lu-labeled analogs revealed a clear superiority of [177Lu]DOTA-r-a-ABA-CPCR4 and [177Lu]DOTA-r-a-ABA-iodoCPCR4 over [177Lu]PentixaTher with respect to tumor uptake (18.3±3.7 and 17.2±2.0 %iD/g, respectively, at 1h p.i. vs 12.4±3.7%iD/g for [177Lu]PentixaTher) as well as activity retention in tumor up to 48h. Especially for [177Lu]DOTA-r-a-ABA-CPCR4 with its low background accumulation, tumor/organ ratios at 48h were 2- to 4-fold higher than those obtained for [177Lu]PentixaTher (except for kidney). Conclusions: The in-depth evaluation of a series of novel CPCR4- and iodoCPCR4 analogs with modified linker structure has yielded reliable structure-activity relationships. It was generally observed that a) AMBA-by-ABA-substitution leads to enhanced ligand internalization, b) the extension of the ABA-linker by two additional amino acids (DOTA-Xaa2-Xaa1-ABA-) provides sufficient linker length to minimize the interaction of the [M3+]DOTA-chelate with the receptor, and that c) introduction of a cationic side chain (Xaa2) greatly enhances receptor affinity of the constructs, obliterating the necessity for Tyr1-iodination of the pentapeptide core to maintain high receptor affinity (such as in [177Lu]PentixaTher). As a result, [177Lu]DOTA-r-a-ABA-CPCR4 has emerged from this study as a powerful second-generation therapeutic CXCR4 ligand with greatly improved targeting efficiency and tumor retention and will be further evaluated in preclinical and clinical CXCR4-targeted dosimetry and RLT studies.
Collapse
|
24
|
Li N, Wang YR, Tian XQ, Lin L, Liang SY, Li QY, Fei X, Tang J, Luo YK. Potential value of three-dimensional ultrasonography in diagnosis of diabetic nephropathy in Chinese diabetic population with kidney injury. BMC Nephrol 2020; 21:243. [PMID: 32600283 PMCID: PMC7325142 DOI: 10.1186/s12882-020-01902-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 06/22/2020] [Indexed: 11/10/2022] Open
Abstract
Background To explore the potential value of three-dimensional ultrasonography (3DUS) and contrast-enhanced ultrasound (CEUS) in the diagnosis of diabetic nephropathy (DN) in Chinese diabetic patients with kidney injury. Methods Patients with type 2 diabetes mellitus and kidney injury in our hospital were enrolled, and the clinical characteristics as well as the laboratory data of patients were collected; 3DUS and CEUS were used to evaluate the morphological structure and blood perfusion of kidneys. Eligible patients were categorized into two groups based on renal biopsy results: DN group and non-diabetic renal diseases (NDRD) group. Correlation analysis and logistic regression analysis were applied to identify the risk factors of DN development. Results A total of 115 patients aged from 24 to 78 years old were recruited in the experiment, of which 64 patients (55.65%) and 51 patients (44.35%) were in the DN group and NDRD group, respectively. After correction to CKD stage, BMI and right kidney volume index were retained to identify patients with DN. The ROC of the logistic regression model had an AUC of 0.703 (95% CI: 0.591–0.815) and it was higher than both indicators. Conclusion 3DUS has potential value in the diagnosis of diabetic nephropathy in Chinese diabetic population with kidney injury and may act as an auxiliary diagnosis for DN. More prospective and well-designed studies with larger samples are needed to verify the result.
Collapse
Affiliation(s)
- Nan Li
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Yi-Ru Wang
- Department of Ultrasound, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiao-Qi Tian
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Lin Lin
- Medical School of Chinese PLA, Beijing, 100853, China
| | - Shu-Yuan Liang
- Department of Ultrasound, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Qiu-Yang Li
- Department of Ultrasound, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Xiang Fei
- Department of Ultrasound, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China
| | - Jie Tang
- Medical School of Chinese PLA, Beijing, 100853, China.
| | - Yu-Kun Luo
- Department of Ultrasound, The First Medical Center, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
25
|
Pajenda S, Rasul S, Hacker M, Wagner L, Geist BK. Dynamic 2-deoxy-2[18F] fluoro-D-glucose PET/MRI in human renal allotransplant patients undergoing acute kidney injury. Sci Rep 2020; 10:8270. [PMID: 32427878 PMCID: PMC7237443 DOI: 10.1038/s41598-020-65267-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 04/29/2020] [Indexed: 12/11/2022] Open
Abstract
Patients after solid organ kidney transplantation (KTX) often suffer from acute kidney injury (AKI). Parameters as serum creatinine indicate a loss of kidney function, although no distinction of the cause and prognosis can be made. Imaging tools measuring kidney function have not been widely in clinical use. In this observational study we evaluated 2-deoxy-2[18F] fluoro-D-glucose (FDG) PET/MRI in thirteen patients after KTX with AKI as a functional assessment of the graft. Twenty-four healthy volunteers served as control. General kidney performance (GKP), initial flow (IF) and renal response function (RF) were calculated by standardized uptake values (SUV) and time activity curves (TAC). The GKP measured for the total kidney and medulla was significantly higher in healthy patients compared to patients after KTX (p = 0.0002 and p = 0.0004, respectively), but no difference was found for the GKP of the cortex (p = 0.59). The IF in KTX patients correlated with renal recovery, defined as change in serum creatinine 10 days after PET/MRI (r = 0.80, p = 0.001). With regard to the RF, a negative correlation for tubular damage was found (r = -0.74, p = 0.004). In conclusion, parameters obtained from FDG PET/MRI showed a possible predictive feature for renal recovery in KTX patients undergoing AKI.
Collapse
Affiliation(s)
- Sahra Pajenda
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna. Waehringer Guertel 18-20, 1090, Vienna, Austria.
| | - Sazan Rasul
- Department of Biomedical Imaging and Image- Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna. Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Marcus Hacker
- Department of Biomedical Imaging and Image- Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna. Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Ludwig Wagner
- Department of Medicine III, Division of Nephrology and Dialysis, Medical University of Vienna. Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Barbara Katharina Geist
- Department of Biomedical Imaging and Image- Guided Therapy, Division of Nuclear Medicine, Medical University of Vienna. Waehringer Guertel 18-20, 1090, Vienna, Austria
| |
Collapse
|
26
|
Pulmonary Cryptococcosis Accidentally Detected by 68Ga-Pentixafor PET/CT in a Patient With Multiple Myeloma. Clin Nucl Med 2020; 45:423-425. [DOI: 10.1097/rlu.0000000000003004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
27
|
Abstract
This review discusses nuclear imaging of inflammation using molecular probes beyond fluoro-d-glucose, is structured by cellular targets, and focuses on those tracers that have been successfully applied clinically.
Collapse
Affiliation(s)
- Malte Kircher
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Augsburg, Stenglinstr. 2, Würzburg 86156, Germany.
| |
Collapse
|
28
|
Tan S, Yang H, Xue S, Qiao J, Salarian M, Hekmatyar K, Meng Y, Mukkavilli R, Pu F, Odubade OY, Harris W, Hai Y, Yushak ML, Morales-Tirado VM, Mittal P, Sun PZ, Lawson D, Grossniklaus HE, Yang JJ. Chemokine receptor 4 targeted protein MRI contrast agent for early detection of liver metastases. SCIENCE ADVANCES 2020; 6:eaav7504. [PMID: 32083172 PMCID: PMC7007242 DOI: 10.1126/sciadv.aav7504] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2018] [Accepted: 11/22/2019] [Indexed: 05/22/2023]
Abstract
Liver metastases often progress from primary cancers including uveal melanoma (UM), breast, and colon cancer. Molecular biomarker imaging is a new non-invasive approach for detecting early stage tumors. Here, we report the elevated expression of chemokine receptor 4 (CXCR4) in liver metastases in UM patients and metastatic UM mouse models, and development of a CXCR4-targeted MRI contrast agent, ProCA32.CXCR4, for sensitive MRI detection of UM liver metastases. ProCA32.CXCR4 exhibits high relaxivities (r 1 = 30.9 mM-1 s-1, r 2 = 43.2 mM-1 s-1, 1.5 T; r 1 = 23.5 mM-1 s-1, r 2 = 98.6 mM-1 s-1, 7.0 T), strong CXCR4 binding (K d = 1.10 ± 0.18 μM), CXCR4 molecular imaging capability in metastatic and intrahepatic xenotransplantation UM mouse models. ProCA32.CXCR4 enables detecting UM liver metastases as small as 0.1 mm3. Further development of the CXCR4-targeted imaging agent should have strong translation potential for early detection, surveillance, and treatment stratification of liver metastases patients.
Collapse
Affiliation(s)
- Shanshan Tan
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Hua Yang
- Department of Ophthalmology, Emory University, Atlanta, GA 30322, USA
| | - Shenghui Xue
- InLighta Biosciences LLC, Atlanta, GA 30303, USA
| | - Jingjuan Qiao
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Mani Salarian
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | - Khan Hekmatyar
- Bioimaging Research Center, University of Georgia, Athens, GA 30602, USA
| | - Yuguang Meng
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - Rao Mukkavilli
- Department of Biology, Georgia State University, Atlanta, GA 30303, USA
| | - Fan Pu
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
| | | | - Wayne Harris
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Yan Hai
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA 30303, USA
| | - Melinda L. Yushak
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | | | - Pardeep Mittal
- Department of Radiology and Imaging, Augusta University, Augusta, GA 30912, USA
| | - Phillip Z. Sun
- Yerkes National Primate Research Center, Emory University, Atlanta, GA 30329, USA
| | - David Lawson
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | | | - Jenny J. Yang
- Department of Chemistry, Georgia State University, Atlanta, GA 30303, USA
- Center for Diagnostics and Therapeutics, Georgia State University, Atlanta, GA 30303, USA
- Corresponding author.
| |
Collapse
|
29
|
Schutter R, Lantinga VA, Borra RJH, Moers C. MRI for diagnosis of post-renal transplant complications: current state-of-the-art and future perspectives. MAGNETIC RESONANCE MATERIALS IN PHYSICS BIOLOGY AND MEDICINE 2019; 33:49-61. [PMID: 31879853 DOI: 10.1007/s10334-019-00813-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 10/27/2019] [Accepted: 11/30/2019] [Indexed: 02/07/2023]
Abstract
Kidney transplantation has developed into a widespread procedure to treat end stage renal failure, with transplantation results improving over the years. Postoperative complications have decreased over the past decades, but are still an important cause of morbidity and mortality. Early accurate diagnosis and treatment is the key to prevent renal allograft impairment or even graft loss. Ideally, a diagnostic tool should be able to detect post-transplant renal dysfunction, differentiate between the different causes and monitor renal function during and after therapeutic interventions. Non-invasive imaging modalities for diagnostic purposes show promising results. Magnetic resonance imaging (MRI) techniques have a number of advantages, such as the lack of ionizing radiation and the possibility to obtain relevant tissue information without contrast, reducing the risk of contrast-induced nephrotoxicity. However, most techniques still lack the specificity to distinguish different types of parenchymal diseases. Despite some promising outcomes, MRI is still barely used in the post-transplantation diagnostic process. The aim of this review is to survey the current literature on the relevance and clinical applicability of diagnostic MRI modalities for the detection of various types of complications after kidney transplantation.
Collapse
Affiliation(s)
- Rianne Schutter
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands.
| | - Veerle A Lantinga
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Ronald J H Borra
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Cyril Moers
- University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| |
Collapse
|
30
|
Adlere I, Caspar B, Arimont M, Dekkers S, Visser K, Stuijt J, de Graaf C, Stocks M, Kellam B, Briddon S, Wijtmans M, de Esch I, Hill S, Leurs R. Modulators of CXCR4 and CXCR7/ACKR3 Function. Mol Pharmacol 2019; 96:737-752. [PMID: 31548340 DOI: 10.1124/mol.119.117663] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 09/14/2019] [Indexed: 02/14/2025] Open
Abstract
The two G protein-coupled receptors (GPCRs) C-X-C chemokine receptor type 4 (CXCR4) and atypical chemokine receptor 3 (ACKR3) are part of the class A chemokine GPCR family and represent important drug targets for human immunodeficiency virus (HIV) infection, cancer, and inflammation diseases. CXCR4 is one of only three chemokine receptors with a US Food and Drug Administration approved therapeutic agent, the small-molecule modulator AMD3100. In this review, known modulators of the two receptors are discussed in detail. Initially, the structural relationship between receptors and ligands is reviewed on the basis of common structural motifs and available crystal structures. To date, no atypical chemokine receptor has been crystallized, which makes ligand design and predictions for these receptors more difficult. Next, the selectivity, receptor activation, and the resulting ligand-induced signaling output of chemokines and other peptide ligands are reviewed. Binding of pepducins, a class of lipid-peptides whose basis is the internal loop of a GPCR, to CXCR4 is also discussed. Finally, small-molecule modulators of CXCR4 and ACKR3 are reviewed. These modulators have led to the development of radio- and fluorescently labeled tool compounds, enabling the visualization of ligand binding and receptor characterization both in vitro and in vivo. SIGNIFICANCE STATEMENT: To investigate the pharmacological modulation of CXCR4 and ACKR3, significant effort has been focused on the discovery and development of a range of ligands, including small-molecule modulators, pepducins, and synthetic peptides. Imaging tools, such as fluorescent probes, also play a pivotal role in the field of drug discovery. This review aims to provide an overview of the aforementioned modulators that facilitate the study of CXCR4 and ACKR3 receptors.
Collapse
Affiliation(s)
- Ilze Adlere
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Birgit Caspar
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Marta Arimont
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Sebastian Dekkers
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Kirsten Visser
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Jeffrey Stuijt
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Chris de Graaf
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Michael Stocks
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Barrie Kellam
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Stephen Briddon
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Maikel Wijtmans
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Iwan de Esch
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Stephen Hill
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| | - Rob Leurs
- Griffin Discoveries BV, Amsterdam, The Netherlands (I.A., I.E., R.L.); Division of Physiology, Pharmacology and Neuroscience, School of Life Sciences (B.C., S.B., S.H.) and School of Pharmacy (S.D., M.S., B.K.), University of Nottingham, Nottingham, United Kingdom; Centre of Membrane Proteins and Receptors (COMPARE), Universities of Birmingham and Nottingham, The Midlands, United Kingdom (B.C., S.D., B.K., S.B., S.H.); Division of Medicinal Chemistry, Amsterdam Institute for Molecules, Medicines and Systems, Faculty of Science, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands (M.A., K.V., J.S., C.G., M.W., I.E., R.L.); and Sosei Heptares, Cambridge, United Kingdom (C.G.)
| |
Collapse
|
31
|
Pan Q, Cao X, Luo Y, Li J, Feng J, Li F. Chemokine receptor-4 targeted PET/CT with 68Ga-Pentixafor in assessment of newly diagnosed multiple myeloma: comparison to 18F-FDG PET/CT. Eur J Nucl Med Mol Imaging 2019; 47:537-546. [PMID: 31776631 DOI: 10.1007/s00259-019-04605-z] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 11/07/2019] [Indexed: 01/07/2023]
Abstract
PURPOSE 18F-FDG PET/CT has some limitations in the evaluation of multiple myeloma (MM). Since chemokine receptor-4 is overexpressed in MM, we perform a prospective cohort study to compare the performance of 68Ga-Pentixafor and 18F-FDG PET/CT in newly diagnosed MM. METHODS Thirty patients with newly diagnosed MM were recruited. All patients underwent 68Ga-Pentixafor and18F-FDG PET/CT within 1 week after enrollment. A positive PET/CT was defined as the presence of focal PET-positive lesions in bone marrow or diffuse bone marrow patterns (uptake > liver). Bone marrow uptake values in 68Ga-Pentixafor and18F-FDG PET/CT (total bone marrow glycolysis [TBmGFDG], total bone marrow uptake with 68Ga-Pentixafor [TBmUCXCR4], total bone marrow volume [TBmV], SUVmean, and SUVmax) were obtained by drawing total bone marrow volume of interest on PET/CT. The positive rates of the PET/CT scans were statistically compared, and the correlation between quantitative bone marrow uptake values and clinical characteristics, laboratory findings, and staging was analyzed. RESULTS 68Ga-Pentixafor PET/CT had a higher positive rate than 18F-FDG PET/CT in recruited patients (93.3 vs. 53.3%, p = 0.0005). In quantitative analysis, bone marrow uptake values in 68Ga-Pentixafor (TBmUCXCR4, SUVmax, and SUVmean) were positively correlated with end organ damage, staging, and laboratory biomarkers related to tumor burden including serum β2-microglobulin, serum free light chain, and 24-h urine light chain (p < 0.05). In 18F-FDG PET/CT, only the SUVmean of total bone marrow was positively correlated with serum free light chain and 24-h urine light chain (p < 0.05). CONCLUSIONS 68Ga-Pentixafor PET/CT is promising in assessment of newly diagnosed MM. TRIAL REGISTRATION NUMBER NCT03436342.
Collapse
Affiliation(s)
- Qingqing Pan
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China
| | - Xinxin Cao
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China
| | - Yaping Luo
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China. .,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China.
| | - Jian Li
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China
| | - Jun Feng
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China
| | - Fang Li
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Wangfujing, Dongcheng District, 100730, Beijing, People's Republic of China
| |
Collapse
|
32
|
Luo Y, Cao X, Pan Q, Li J, Feng J, Li F. 68Ga-Pentixafor PET/CT for Imaging of Chemokine Receptor 4 Expression in Waldenström Macroglobulinemia/Lymphoplasmacytic Lymphoma: Comparison to 18F-FDG PET/CT. J Nucl Med 2019; 60:1724-1729. [PMID: 31101745 DOI: 10.2967/jnumed.119.226134] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2019] [Accepted: 05/07/2019] [Indexed: 12/14/2022] Open
Abstract
18F-FDG PET/CT has some limitations in the evaluation of Waldenström macroglobulinemia/lymphoplasmacytic lymphoma (WM/LPL), an indolent B-cell lymphoma that primarily involves the bone marrow. Because there is a high level of chemokine receptor 4 expression in the B cells of WM/LPL patients, we performed a prospective cohort study to evaluate the performance of 68Ga-pentixafor, which targets chemokine receptor 4 in WM/LPL, and to compare it with the performance of 18F-FDG. Methods: Seventeen patients with WM/LPL were recruited. All patients underwent both 68Ga-pentixafor PET/CT and 18F-FDG PET/CT. A positive PET/CT result was defined as the presence of focal lesions with positive PET results or diffuse bone marrow patterns (uptake > liver). The rates of positive results for PET/CT scans of bone marrow, lymph nodes, and other extramedullary involvement were statistically compared. Results: 68Ga-pentixafor PET/CT had a higher rate of positive results than 18F-FDG PET/CT (100% vs. 58.8%; P = 0.023) in the recruited WM/LPL patients. The sensitivities of 68Ga-pentixafor PET/CT and 18F-FDG PET/CT for detecting bone marrow involvement were 94.1% and 58.8%, respectively (P = 0.077). In terms of detecting lymph node involvement, 68Ga-pentixafor PET/CT had a significantly higher rate of positive results than 18F-FDG PET/CT (76.5% vs. 11.8%; P = 0.003). In addition, 68Ga-pentixafor detected more paramedullary and central nervous system involvement than 18F-FDG. Conclusion: 68Ga-pentixafor might be a promising imaging agent for the assessment of WM/LPL.
Collapse
Affiliation(s)
- Yaping Luo
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China; and
| | - Xinxin Cao
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Qingqing Pan
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, People's Republic of China.,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China; and
| | - Jian Li
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Jun Feng
- Department of Hematology, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, People's Republic of China
| | - Fang Li
- Department of Nuclear Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College Hospital, Beijing, People's Republic of China .,Beijing Key Laboratory of Molecular Targeted Diagnosis and Therapy in Nuclear Medicine, Beijing, People's Republic of China; and
| |
Collapse
|
33
|
Luo J, Chen J, Sun Y, Zhou H, Xu K, Huang F, Huang P. Quantitative contrast-enhanced ultrasound of renal perfusion: a technology for the assessment of early diabetic nephropathy in cynomolgus macaques with type 2 diabetes mellitus. Abdom Radiol (NY) 2019; 44:1850-1857. [PMID: 30694370 DOI: 10.1007/s00261-019-01908-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
PURPOSE The aim of this study was to investigate the effectiveness of contrast-enhanced ultrasound (CEUS) in predicting early nephropathy in cynomolgus macaques with spontaneous type 2 diabetes mellitus (T2DM). METHODS Six cynomolgus macaques with spontaneous T2DM and six normal cynomolgus macaques (Group 1) were included in this study. The time-intensity curve was used to obtain parameters such as peak values, red blood volume (RBV), red blood flow (RBF), time to peak (TTP), and mean transit time (MTT). Biopsy renal tissue samples were assessed histopathologically. Six cynomolgus macaques with spontaneous T2DM were subgrouped into T2DM without nephropathy group (Group 2) and T2DM with nephropathy group (Group 3) based on histopathological findings. RESULTS Peak value had the largest area under the curve comparing with RBF, RBV, TTP, MTT. The sensitivity and specificity of peak value with cut-off value of 38.65 dB for the diagnosis of DN were 98.3% and 83%, respectively. Peak value, RBV, and RBF in Group 3 was significantly decreased compared with Group 1 and Group 2 (P = 0.000, x2 = 23.99; P = 0.003, x2 = 9.14; P = 0.02, x2 = 5.14). CONCLUSIONS The perfusion parameter of peak value in CEUS might be useful in predicting early diabetic nephropathy in spontaneous T2DM cynomolgus macaques.
Collapse
|
34
|
Derlin T, Hueper K. CXCR4-targeted therapy in breast cancer. Lancet Oncol 2019; 19:e370. [PMID: 30102220 DOI: 10.1016/s1470-2045(18)30480-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2018] [Accepted: 06/18/2018] [Indexed: 11/25/2022]
Affiliation(s)
- Thorsten Derlin
- Department of Nuclear Medicine, Hannover Medical School, Hannover D-30625, Germany.
| | - Katja Hueper
- Department of Radiology, Hannover Medical School, Hannover D-30625, Germany
| |
Collapse
|
35
|
Abstract
Kidney diseases can be caused by a wide range of genetic, hemodynamic, toxic, infectious, and autoimmune factors. The diagnosis of kidney disease usually involves the biochemical analysis of serum and blood, but these tests are often insufficiently sensitive or specific to make a definitive diagnosis. Although radiologic imaging currently has a limited role in the evaluation of most kidney diseases, several new imaging methods hold great promise for improving our ability to non-invasively detect structural, functional, and molecular changes within the kidney. New methods, such as dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) and blood oxygen level-dependent (BOLD) MRI, allow functional imaging of the kidney. The use of novel contrast agents, such as microbubbles and nanoparticles, allows the detection of specific molecules in the kidney. These methods could greatly advance our ability to diagnose disease and also to safely monitor patients over time. This could improve the care of individual patients, and it could also facilitate the evaluation of new treatment strategies.
Collapse
Affiliation(s)
- Joshua Thurman
- Department of Medicine, University of Colorado School of Medicine, Aurora, CO, USA
| | - Faikah Gueler
- Department of Nephrology, Hannover Medical School, Hannover, Germany
| |
Collapse
|
36
|
Kircher M, Herhaus P, Schottelius M, Buck AK, Werner RA, Wester HJ, Keller U, Lapa C. CXCR4-directed theranostics in oncology and inflammation. Ann Nucl Med 2018; 32:503-511. [PMID: 30105558 PMCID: PMC6182637 DOI: 10.1007/s12149-018-1290-8] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2018] [Accepted: 08/08/2018] [Indexed: 12/17/2022]
Abstract
Given its prominent role in inflammation and cancer biology, the C-X-C motif chemokine receptor 4 (CXCR4) has gained a lot of attention in the recent years. This review gives a short overview of the physiology and pathology of chemokines and chemokine receptors and then focuses on the current experience of targeting CXCR4, using radiolabeled receptor ligands suitable for positron emission tomography (PET) imaging, in both hematologic and solid malignancy as well as in inflammatory conditions. Additionally, CXCR4-directed endoradiotherapy (ERT) as a new treatment option is discussed.
Collapse
Affiliation(s)
- Malte Kircher
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Peter Herhaus
- Internal Medicine III, Hematology and Medical Oncology, Technische Universität München, Munich, Germany
| | - Margret Schottelius
- Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany
| | - Andreas K Buck
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
| | - Rudolf A Werner
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany
- Division of Nuclear Medicine and Molecular Imaging, Johns Hopkins University School of Medicine, Baltimore, USA
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technische Universität München, Munich, Germany
| | - Ulrich Keller
- Internal Medicine III, Hematology and Medical Oncology, Technische Universität München, Munich, Germany
| | - Constantin Lapa
- Department of Nuclear Medicine, University Hospital Würzburg, Oberdürrbacher Str. 6, 97080, Würzburg, Germany.
| |
Collapse
|
37
|
Caroli A, Schneider M, Friedli I, Ljimani A, De Seigneux S, Boor P, Gullapudi L, Kazmi I, Mendichovszky IA, Notohamiprodjo M, Selby NM, Thoeny HC, Grenier N, Vallée JP. Diffusion-weighted magnetic resonance imaging to assess diffuse renal pathology: a systematic review and statement paper. Nephrol Dial Transplant 2018; 33:ii29-ii40. [PMID: 30137580 PMCID: PMC6106641 DOI: 10.1093/ndt/gfy163] [Citation(s) in RCA: 120] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/02/2018] [Indexed: 12/26/2022] Open
Abstract
Diffusion-weighted magnetic resonance imaging (DWI) is a non-invasive method sensitive to local water motion in the tissue. As a tool to probe the microstructure, including the presence and potentially the degree of renal fibrosis, DWI has the potential to become an effective imaging biomarker. The aim of this review is to discuss the current status of renal DWI in diffuse renal diseases. DWI biomarkers can be classified in the following three main categories: (i) the apparent diffusion coefficient-an overall measure of water diffusion and microcirculation in the tissue; (ii) true diffusion, pseudodiffusion and flowing fraction-providing separate information on diffusion and perfusion or tubular flow; and (iii) fractional anisotropy-measuring the microstructural orientation. An overview of human studies applying renal DWI in diffuse pathologies is given, demonstrating not only the feasibility and intra-study reproducibility of DWI but also highlighting the need for standardization of methods, additional validation and qualification. The current and future role of renal DWI in clinical practice is reviewed, emphasizing its potential as a surrogate and monitoring biomarker for interstitial fibrosis in chronic kidney disease, as well as a surrogate biomarker for the inflammation in acute kidney diseases that may impact patient selection for renal biopsy in acute graft rejection. As part of the international COST (European Cooperation in Science and Technology) action PARENCHIMA (Magnetic Resonance Imaging Biomarkers for Chronic Kidney Disease), aimed at eliminating the barriers to the clinical use of functional renal magnetic resonance imaging, this article provides practical recommendations for future design of clinical studies and the use of renal DWI in clinical practice.
Collapse
Affiliation(s)
- Anna Caroli
- Medical Imaging Unit, Bioengineering Department, IRCCS Istituto di Ricerche Farmacologiche Mario Negri, Bergamo, Italy
| | - Moritz Schneider
- Department of Radiology, Ludwig-Maximilians-University Hospital Munich, Munich, Germany
- Comprehensive Pneumology Center, German Center for Lung Research, Munich, Germany
| | - Iris Friedli
- Division of Radiology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Alexandra Ljimani
- Department of Diagnostic and Interventional Radiology, Medical Faculty, University Dusseldorf, Dusseldorf, Germany
| | - Sophie De Seigneux
- Service and Laboratory of Nephrology, Department of Internal Medicine Specialties and Department of Physiology and Metabolism, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| | - Peter Boor
- Institute of Pathology and Division of Nephrology, RWTH University of Aachen, Aachen, Germany
| | - Latha Gullapudi
- Centre for Kidney Research and Innovation, University of Nottingham, Nottingham, UK
| | - Isma Kazmi
- Centre for Kidney Research and Innovation, University of Nottingham, Nottingham, UK
| | - Iosif A Mendichovszky
- Department of Radiology, Cambridge University Hospitals NHS Foundation Trust, Addenbrooke’s Hospital, Cambridge, UK
| | | | - Nicholas M Selby
- Centre for Kidney Research and Innovation, University of Nottingham, Nottingham, UK
| | - Harriet C Thoeny
- Department of Diagnostic, Pediatric, and Interventional Radiology, Inselspital University Hospital, Bern, Switzerland
| | - Nicolas Grenier
- Service d'Imagerie Diagnostique et Interventionnelle de l'Adulte, Centre Hospitalier Universitaire de Bordeaux, Bordeaux, France
| | - Jean-Paul Vallée
- Division of Radiology, Geneva University Hospitals, University of Geneva, Geneva, Switzerland
| |
Collapse
|
38
|
Chemokine receptor - Directed imaging and therapy. Methods 2017; 130:63-71. [PMID: 28916148 DOI: 10.1016/j.ymeth.2017.09.002] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2017] [Revised: 09/05/2017] [Accepted: 09/09/2017] [Indexed: 12/29/2022] Open
Abstract
The C-X-C chemokine receptor 4 (CXCR4) and its natural ligand CXCL12 are key factors in the process of cell migration, homing of hematopoietic stem cells to the bone marrow, and represent important mediators of angiogenesis and cell proliferation. The CXCR4/CXCL12 interplay can be disrupted by CXCR4 antagonists such as Plerixafor which are already in daily clinical use, i.e. for mobilization and subsequent harvesting of hematopoietic progenitor cells and stem cell transplantation. In a pathological condition, involvement in the process of metastasis and homing of cancer cells to a protective niche has been described, making CXCR4 an attractive target for imaging and treatment of malignant diseases. Recently, radiolabeled analogs of CXCR4 antagonists (e.g., [68Ga]Pentixafor) have been introduced which can be used for non-invasive imaging of CXCR4 expression in animal models and humans using positron emission tomography. In addition, beta emitter-labeled antagonists (i.e., [177Lu]/[90Y]Pentixather) have been used in small patient cohorts for treatment of hematological neoplasms such as lymphoma, multiple myeloma and acute myeloid leukemia. This review reports on current imaging protocols for CXCR4-directed positron emission tomography in preclinical models and in humans. Furthermore, a theranostic approach using beta emitter-labeled antagonists is highlighted. Molecular imaging of the CXCR4/CXCL12 axis can contribute to further understand the process of metastatic spread and the intra-/interindividual heterogeneity of tumors. In addition, CXCR4 directed imaging allows tracking of activated, CXCR4+ immune cells. This allows for watching inflammatory processes, thus contributing to enlighten the role of the immune system in a variety of cardiovascular and neurological diseases.
Collapse
|