1
|
Huang J, Chen X, Jiang Y, Zhang C, He S, Wang H, Pu K. Renal clearable polyfluorophore nanosensors for early diagnosis of cancer and allograft rejection. NATURE MATERIALS 2022; 21:598-607. [PMID: 35422505 DOI: 10.1038/s41563-022-01224-2] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Academic Contribution Register] [Received: 05/18/2021] [Accepted: 02/21/2022] [Indexed: 06/14/2023]
Abstract
Optical nanoparticles are promising diagnostic tools; however, their shallow optical imaging depth and slow clearance from the body have impeded their use for in vivo disease detection. To address these limitations, we develop activatable polyfluorophore nanosensors with biomarker-triggered nanoparticle-to-molecule pharmacokinetic conversion and near-infrared fluorogenic turn-on response. Activatable polyfluorophore nanosensors can accumulate at the disease site and react with disease-associated proteases to undergo in situ enzyme-catalysed depolymerization. This disease-specific interaction liberates renal-clearable fluorogenic fragments from activatable polyfluorophore nanosensors for non-invasive longitudinal urinalysis and outperforms the gold standard blood and urine assays, providing a level of sensitivity and specificity comparable to those of invasive biopsy and flow cytometry analysis. In rodent models, activatable polyfluorophore nanosensors enable ultrasensitive detection of tumours (1.6 mm diameter) and early diagnosis of acute liver allograft rejection. We anticipate that our modular nanosensor platform may be applied for early diagnosis of a range of diseases via a simple urine test.
Collapse
Affiliation(s)
- Jiaguo Huang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Xiaona Chen
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, P. R. China
| | - Yuyan Jiang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Chi Zhang
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Shasha He
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore
| | - Hangxiang Wang
- The First Affiliated Hospital, Zhejiang University School of Medicine; NHC Key Laboratory of Combined Multi-Organ Transplantation; Key Laboratory of Organ Transplantation, Research Center for Diagnosis and Treatment of Hepatobiliary Diseases, Zhejiang Province, Hangzhou, P. R. China
| | - Kanyi Pu
- School of Chemical and Biomedical Engineering, Nanyang Technological University, Singapore, Singapore.
| |
Collapse
|
2
|
Abstract
One major characteristic of programmed cell death (apoptosis) results in the increased expression of phosphatidylserine (PS) on the outer membrane of dying cells. Consequently, PS represents an excellent target for non-invasive imaging of apoptosis by single-photon emission computed tomography (SPECT) and positron emission tomography (PET). Annexin V is a 36 kDa protein which binds with high affinity to PS in the presence of Ca2+ ions. This makes radiolabeled annexins valuable apoptosis imaging agents for clinical and biomedical research applications for monitoring apoptosis in vivo. However, the use of radiolabeled annexin V for in vivo imaging of cell death has been met with a variety of challenges which have prevented its translation into the clinic. These difficulties include: complicated and time-consuming radiolabeling procedures, sub-optimal biodistribution, inadequate pharmacokinetics leading to poor tumour-to-blood contrast ratios, reliance upon Ca2+ concentrations in vivo, low tumor tissue penetration, and an incomplete understanding of what constitutes the best imaging protocol following induction of apoptosis. Therefore, new concepts and improved strategies for the development of PS-binding radiotracers are needed. Radiolabeled PS-binding peptides and various Zn(II) complexes as phosphate chemosensors offer an innovative strategy for radionuclide-based molecular imaging of apoptosis with PET and SPECT. Radiolabeled peptides and Zn(II) complexes provide several advantages over annexin V including better pharmacokinetics due to their smaller size, better availability, simpler synthesis and radiolabeling strategies as well as facilitated tissue penetration due to their smaller size and faster blood clearance profile allowing for optimized image contrast. In addition, peptides can be structurally modified to improve metabolic stability along with other pharmacokinetic and pharmacodynamic properties. The present review will summarize the current status of radiolabeled annexins, peptides and Zn(II) complexes developed as radiotracers for imaging apoptosis through targeting PS utilizing PET and SPECT imaging.
Collapse
|
3
|
Aghighi M, Pisani L, Theruvath AJ, Muehe AM, Donig J, Khan R, Holdsworth SJ, Kambham N, Concepcion W, Grimm PC, Daldrup-Link HE. Ferumoxytol Is Not Retained in Kidney Allografts in Patients Undergoing Acute Rejection. Mol Imaging Biol 2018; 20:139-149. [PMID: 28411307 PMCID: PMC6391060 DOI: 10.1007/s11307-017-1084-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/11/2022]
Abstract
PURPOSE To evaluate whether ultrasmall superparamagnetic iron oxide nanoparticle (USPIO)-enhanced magnetic resonance imaging (MRI) can detect allograft rejection in pediatric kidney transplant patients. PROCEDURES The USPIO ferumoxytol has a long blood half-life and is phagocytosed by macrophages. In an IRB-approved single-center prospective clinical trial, 26 pediatric patients and adolescents (age 10-26 years) with acute allograft rejection (n = 5), non-rejecting allografts (n = 13), and normal native kidneys (n = 8) underwent multi-echo T2* fast spoiled gradient-echo (FSPGR) MRI after intravenous injection (p.i.) of 5 mg Fe/kg ferumoxytol. T2* relaxation times at 4 h p.i. (perfusion phase) and more than 20 h p.i. (macrophage phase) were compared with biopsy results. The presence of rejection was assessed using the Banff criteria, and the prevalence of macrophages on CD163 immunostains was determined based on a semi-quantitative scoring system. MRI and histology data were compared among patient groups using t tests, analysis of variance, and regression analyses with a significance threshold of p < 0.05. RESULTS At 4 h p.i., mean T2* values were 6.6 ± 1.5 ms for native kidneys and 3.9 ms for one allograft undergoing acute immune rejection. Surprisingly, at 20-24 h p.i., one rejecting allograft showed significantly prolonged T2* relaxation times (37.0 ms) compared to native kidneys (6.3 ± 1.7 ms) and non-rejecting allografts (7.6 ± 0.1 ms). Likewise, three additional rejecting allografts showed significantly prolonged T2* relaxation times compared to non-rejecting allografts at later post-contrast time points, 25-97 h p.i. (p = 0.008). Histological analysis revealed edema and compressed microvessels in biopsies of rejecting allografts. Allografts with and without rejection showed insignificant differences in macrophage content on histopathology (p = 0.44). CONCLUSION After ferumoxytol administration, renal allografts undergoing acute rejection show prolonged T2* values compared to non-rejecting allografts. Since histology revealed no significant differences in macrophage content, the increasing T2* value is likely due to the combined effect of reduced perfusion and increased edema in rejecting allografts.
Collapse
Affiliation(s)
- Maryam Aghighi
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Laura Pisani
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Ashok J Theruvath
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Anne M Muehe
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Jessica Donig
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Ramsha Khan
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Samantha J Holdsworth
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA
| | - Neeraja Kambham
- Department of Pathology, Stanford University, Stanford, CA, USA
| | | | - Paul C Grimm
- Department of Pediatrics, Stanford University, Stanford, CA, USA
| | - Heike E Daldrup-Link
- Department of Radiology, Pediatric Molecular Imaging in the Molecular Imaging Program at Stanford (@PedsMIPS), Lucile Packard Children's Hospital, Stanford University School of Medicine, 725 Welch Road, Stanford, 94305, CA, USA.
- Department of Pediatrics, Lucile Packard Children's Hospital, Stanford School of Medicine, 725 Welch Rd, Stanford, CA, 94305, USA.
| |
Collapse
|
4
|
Chen DL, Zhou D, Chu W, Herrbrich P, Engle JT, Griffin E, Jones LA, Rothfuss JM, Geraci M, Hotchkiss RS, Mach RH. Radiolabeled isatin binding to caspase-3 activation induced by anti-Fas antibody. Nucl Med Biol 2011; 39:137-44. [PMID: 22033021 DOI: 10.1016/j.nucmedbio.2011.08.001] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/27/2011] [Revised: 07/20/2011] [Accepted: 08/01/2011] [Indexed: 11/30/2022]
Abstract
INTRODUCTION Noninvasive imaging methods that can distinguish apoptosis from necrosis may be useful in furthering our understanding of diseases characterized by apoptotic dysregulation as well as aiding drug development targeting apoptotic pathways. We evaluated the ability of radiolabeled isatins to quantify caspase-3 activity induced by the activation of the extrinsic apoptotic pathway by the anti-Fas antibody in mice. METHODS The behavior of three different radiolabeled isatins ([(18)F]WC-II-89, [(18)F]WC-IV-3 and [(11)C]WC-98) was characterized in mice with and without anti-Fas antibody treatment by microPET imaging and biodistribution studies. The activity of [(18)F]WC-II-89 was also compared with [(99m)Tc]mebrofenin. The effect of pan-caspase inhibition with quinolyl-valyl-O-methylaspartyl-[2,6-difluorophenoxy]-methyl ketone (Q-VD-OPh) on [(18)F]WC-II-89 uptake was studied. Caspase-3 activity was confirmed by a fluorometric enzyme assay. RESULTS All three tracers behaved similarly in microPET and biodistribution studies. Increased retention of all tracers was observed in the livers of treated animals and several other organs, all of which demonstrated increased caspase-3 enzyme activity; however, impaired hepatobiliary excretion made attribution of these findings to caspase-3 activity difficult. The isatin [(18)F]WC-II-89 was retained at statistically significantly higher levels in the organs after anti-Fas antibody treatment while [(99m)Tc]mebrofenin activity cleared, suggesting specific binding to activated caspase-3, but the magnitude of increased binding was still relatively low. Caspase inhibition with Q-VD-OPh partially blocked [(18)F]WC-II-89 retention but completely blocked caspase-3 enzyme activity in the liver. CONCLUSIONS The radiolabeled isatins appear to bind specifically to caspase-3 in vivo, but their sensitivity is limited. Further optimization is required for these tracers to be useful for clinical applications.
Collapse
Affiliation(s)
- Delphine L Chen
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
5
|
Tekabe Y, Li Q, Luma J, Weisenberger D, Sedlar M, Harja E, Narula J, Johnson LL. Noninvasive monitoring the biology of atherosclerotic plaque development with radiolabeled annexin V and matrix metalloproteinase inhibitor in spontaneous atherosclerotic mice. J Nucl Cardiol 2010; 17:1073-81. [PMID: 20700679 DOI: 10.1007/s12350-010-9276-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 02/22/2010] [Accepted: 07/18/2010] [Indexed: 10/19/2022]
Abstract
OBJECTIVES To compare the ability of (99m)Tc-labeled broad-based matrix metalloproteinase inhibitor (RP805) (MPI) and (99m)Tc-annexin V to identify more advanced atherosclerotic disease in apolipoprotein E-null (apoE(-/-)) mice. BACKGROUND Both MMP expression and apoptotic cell death occur in both early and in advanced atherosclerotic plaques. METHODS Eight 6-9-week-old apoE(-/-) mice, 10 apoE(-/-) mice at 20 weeks, and 12 apoE(-/-) at 40 weeks were injected with both tracers in alternating sequence separated by 48 h, underwent planar imaging and were killed. Radiotracer uptake was quantified from the scans as percent whole body and from tissue as percent injected dose per gram (%ID/g). Quantitative immunohistopathology of the aorta and carotids for macrophages, MMPs, and caspase was performed. RESULTS At 6 weeks, mice showed no tracer uptake in the chest or neck and had minimal lesion. At 20 weeks, uptake of annexin V as %ID was borderline higher than MPI (1.10 ± .48% vs .77 ± .31%, P = .09), between 20 and 40 weeks aortic lesion area increased from 37.4 ± 12.0% to 46.2 ± 7.4% and at 40 weeks MPI was significantly greater than annexin V uptake (1.11 ± .66% vs .70 ± .16%, P = .05). On histology there were greater increases in % MMP-2 and -9 than % caspase positive cells. Carotid uptake of MPI was greater than annexin V at both 20 and 40 weeks (1.25 ± .48% vs .78 ± .25%, P = .02 and 3.70 ± 1.45% vs 2.25 ± .66%, P = .005). The carotid lesion area at 40 weeks was 74 ± 9% with greater % cells positive for MMP's than caspase. %ID/g annexin V correlated significantly with % macrophages and with caspase-3 positive cells and %ID/g MPI correlated significantly with % macrophages and with MMP-2 and -9 positive cells. CONCLUSIONS In apoE(-/-) mice, MMP expression is greater than apoptosis as the disease progresses and MPI may be a better imaging agent for more advanced disease.
Collapse
Affiliation(s)
- Yared Tekabe
- Division of Cardiology, Department of Medicine, Columbia University Medical Center, New York, NY 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Zhuo M, Fujiki M, Wang M, Piard-Ruster K, Wai LE, Wei L, Martinez OM, Krams SM. Identification of the rat NKG2D ligands, RAE1L and RRLT, and their role in allograft rejection. Eur J Immunol 2010; 40:1748-57. [PMID: 20306467 DOI: 10.1002/eji.200939779] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/13/2022]
Abstract
NKG2D is a receptor expressed by NK cells and subsets of T lymphocytes. On NK cells, NKG2D functions as a stimulatory receptor that induces effector functions. We cloned and expressed two rat NKG2D ligands, both members of the RAE1 family, RAE1L and RRLT, and demonstrate that these ligands can induce IFN-gamma secretion and cytotoxicity by rat NK cells. To examine changes in expression of NKG2D and the NKG2D ligands RAE1L and RRLT after transplantation, we used a Dark Agouti (DA)-->Lewis rat model of liver transplantation. NKG2D expression was significantly increased in allogeneic liver grafts by day 7 post-transplant. Ligands of NKG2D, absent in normal liver, were readily detected in both syngeneic and allogeneic liver grafts by day 1 post-transplant. By day 7 post-transplant, hepatocyte RAE1L and RRLT expression was significantly and specifically increased in liver allografts. In contrast to acute rejection that develops in the DA-->Lewis model, transplantation of Lewis livers into DA recipients (Lewis-->DA) results in spontaneous tolerance. Interestingly, expression of RAE1L and RRLT is low in Lewis-->DA liver allografts, but significantly increased in DA-->Lewis liver allografts undergoing rejection. In conclusion, our results suggest that expression of NKG2D ligands may be important in allograft rejection.
Collapse
Affiliation(s)
- Ming Zhuo
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, Stanford, CA 94305-5492, USA
| | | | | | | | | | | | | | | |
Collapse
|
7
|
|
8
|
Flotats A, Carrió I. Radionuclide noninvasive evaluation of heart failure beyond left ventricular function assessment. J Nucl Cardiol 2009; 16:304-15. [PMID: 19247733 DOI: 10.1007/s12350-009-9064-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/20/2009] [Accepted: 01/25/2009] [Indexed: 01/08/2023]
Abstract
The management of patients with heart failure (HF) is challenging and requires the integration of clinical skills and accurate ancillary tests for the correct diagnosis and estimation of individual prognosis. Although the basic characterization of patients with HF is supported primarily by echocardiographic assessment of the left ventricular function, other noninvasive imaging procedures are being developed, including those involved in the processes of myocardial perfusion, metabolism, cellular injury, intersticial dysregulation, and neurohormonal receptor function. Nuclear techniques for molecular imaging of the myocardium may provide valuable insights into the pathophysiology, severity, management (medical/mechanical/surgical), response to treatment, and prognosis of HF patients. This will permit individualized management decisions and hopefully facilitate better clinical outcomes for patients with HF.
Collapse
Affiliation(s)
- Albert Flotats
- Nuclear Medicine Department, Hospital de la Santa Creu i Sant Pau, Universitat Autònoma de Barcelona, Sant Antoni M. Claret, 167, Barcelona, 08025, Spain.
| | | |
Collapse
|
9
|
Kartachova MS, Verheij M, van Eck BL, Hoefnagel CA, Olmos RAV. Radionuclide Imaging of Apoptosis in Malignancies: Promise and Pitfalls of Tc-Hynic-rh-Annexin V Imaging. Clin Med Oncol 2008; 2:319-25. [PMID: 21892293 PMCID: PMC3161632 DOI: 10.4137/cmo.s349] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/17/2022] Open
Abstract
Radionuclide detection of apoptosis with of 99mTc-Hynic-rh-Annexin V scintigraphy is an effective tool for in vivo visualisation and monitoring of apoptosis in various malignant tumour. Early therapy-induced increase of the tumour tracer uptake correlates with favourable outcome, whereas stable or decreased uptake correlates with stable disease or tumour progression. Therefore sequential 99mTc-Hynic-rh-Annexin V scintigraphy could be used to predict therapy outcome on a patient-to-patient basis within 48 hours after the start of treatment. However, moderate tumour-to-background ratio and therapy-induced changes in normal tissues could confound image analysis. To assure accurate interpretation of Annexin V scans, the awareness of the biophysiological and biochemical properties contributing to the tracer distribution is essential. In with manuscript we discuss the patterns of Annexin V tumour uptake and illustrate the most frequent pitfalls associated with Annexin V imaging in correlation with CT and MRI imaging.
Collapse
Affiliation(s)
- M S Kartachova
- Department of Nuclear Medicine, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands
| | | | | | | | | |
Collapse
|
10
|
Kurihara H, Yang DJ, Cristofanilli M, Erwin WD, Yu DF, Kohanim S, Mendez R, Kim EE. Imaging and dosimetry of 99mTc EC annexin V: preliminary clinical study targeting apoptosis in breast tumors. Appl Radiat Isot 2008; 66:1175-82. [PMID: 18308577 DOI: 10.1016/j.apradiso.2008.01.012] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 10/16/2007] [Revised: 01/10/2008] [Accepted: 01/11/2008] [Indexed: 11/18/2022]
Abstract
BACKGROUND Early detection of cellular events is important to predict the outcome of the patients. This study was aimed to use (99m)Tc EC-annexin V to image tumor cells undergoing apoptosis. METHODS In 10 patients with breast cancer, scintigraphic images and dosimetric estimates were obtained after administering (99m)Tc EC-annexin V. RESULTS Nine of the 10 cases showed detectable (99m)Tc EC-annexin V uptake in tumor. Higher values of T/N ratios are associated with patient after treatment. CONCLUSIONS Apoptosis can be quantified using (99m)Tc EC-annexin V.
Collapse
Affiliation(s)
- Hiroaki Kurihara
- Department of Experimental Diagnostic Imaging, The University of Texas M.D. Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Hoebers FJP, Kartachova M, de Bois J, van den Brekel MWM, van Tinteren H, van Herk M, Rasch CRN, Valdés Olmos RA, Verheij M. 99mTc Hynic-rh-Annexin V scintigraphy for in vivo imaging of apoptosis in patients with head and neck cancer treated with chemoradiotherapy. Eur J Nucl Med Mol Imaging 2007; 35:509-18. [PMID: 17994297 PMCID: PMC2275773 DOI: 10.1007/s00259-007-0624-x] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 06/22/2007] [Accepted: 09/28/2007] [Indexed: 11/25/2022]
Abstract
Purpose The purpose of this study was to determine the value of 99mTc Hynic-rh-Annexin-V-Scintigraphy (TAVS), a non-invasive in vivo technique to demonstrate apoptosis in patients with head and neck squamous cell carcinoma. Methods TAVS were performed before and within 48 h after the first course of cisplatin-based chemoradiation. Radiation dose given to the tumour at the time of post-treatment TAVS was 6–8 Gy. Single-photon emission tomography data were co-registered to planning CT scan. Complete sets of these data were available for 13 patients. The radiation dose at post-treatment TAVS was calculated for several regions of interest (ROI): primary tumour, involved lymph nodes and salivary glands. Annexin uptake was determined in each ROI, and the difference between post-treatment and baseline TAVS represented the absolute Annexin uptake: Delta uptake (ΔU). Results In 24 of 26 parotid glands, treatment-induced Annexin uptake was observed. Mean ΔU was significantly correlated with the mean radiation dose given to the parotid glands (r = 0.59, p = 0.002): Glands that received higher doses showed more Annexin uptake. ΔU in primary tumour and pathological lymph nodes showed large inter-patient differences. A high correlation was observed on an inter-patient level (r = 0.71, p = 0.006) between the maximum ΔU in primary tumour and in the lymph nodes. Conclusions Within the dose range of 0–8 Gy, Annexin-V-scintigraphy showed a radiation-dose-dependent uptake in parotid glands, indicative of early apoptosis during treatment. The inter-individual spread in Annexin uptake in primary tumours could not be related to differences in dose or tumour volume, but the Annexin uptake in tumour and lymph nodes were closely correlated. This effect might represent a tumour-specific apoptotic response.
Collapse
Affiliation(s)
- Frank J P Hoebers
- Department of Radiotherapy, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, Amsterdam, The Netherlands,
| | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Sarda-Mantel L, Michel JB, Rouzet F, Martet G, Louedec L, Vanderheyden JL, Hervatin F, Raguin O, Vrigneaud JM, Khaw BA, Le Guludec D. (99m)Tc-annexin V and (111)In-antimyosin antibody uptake in experimental myocardial infarction in rats. Eur J Nucl Med Mol Imaging 2005; 33:239-45. [PMID: 16283183 DOI: 10.1007/s00259-005-1900-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 07/01/2005] [Accepted: 07/04/2005] [Indexed: 11/26/2022]
Abstract
PURPOSE (99m)Tc-annexin V (ANX) allows scintigraphic detection of apoptotic cells via specific binding to exposed phosphatidylserine. In myocardial infarction, apoptosis of myocytes is variable and depends especially on the presence or absence of coronary reperfusion. In this study, ANX uptake in non-reperfused experimental myocardial infarcts was compared with uptake of a marker of myocyte necrosis ((111)In-antimyosin antibodies, AM) and an immunohistochemical marker of apoptosis (Apostain). METHODS The left anterior coronary artery was ligated in 47 Wistar rats, which were then injected with ANX (n=20), AM (n=21) or both (n=6). Myocardial uptake of ANX and AM was determined at 2 h (n=14), 4 h (n=14) and 24 h (n=19) after coronary ligation (CL), by quantitative autoradiography with (n=23) or without (n=24) gamma imaging. Heart-to-lung ratios (HLRs) and infarct-to-remote myocardium activity ratios (INRs) were calculated on the scintigrams and autoradiograms respectively. Cardiac sections were stained with haematoxylin-eosin and Apostain. The above studies were repeated in 12 normal rats. RESULTS All rats with CL showed increased ANX and AM uptake in cardiac areas on scintigrams 24 h after CL, with HLRs higher than in controls: 3.1+/-0.6 versus 1.5+/-0.3 (p=0.001) for ANX and 1.99+/-0.44 versus 1.01+/-0.05 (p<0.0005) for AM. Autoradiography showed intense ANX and AM uptake in infarcts, with comparable topography and INRs at 2 h, 4 h and 24 h after CL (4.6+/-0.9 versus 5.0+/-1.8 at 24 h), while Apostain staining was very low (0.06+/-0.06% of cells). CONCLUSION In this model of persistent CL, we observed increased ANX uptake in injured myocardium, comparable in intensity, topography and kinetics to that of AM. There was only minimal Apostain staining in the same areas.
Collapse
Affiliation(s)
- Laure Sarda-Mantel
- EA 3512, Nuclear Medicine Department, Bichat Hospital AP-HP, Paris, France.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Toretsky J, Levenson A, Weinberg IN, Tait JF, Uren A, Mease RC. Preparation of F-18 labeled annexin V: a potential PET radiopharmaceutical for imaging cell death. Nucl Med Biol 2005; 31:747-52. [PMID: 15246365 DOI: 10.1016/j.nucmedbio.2004.02.007] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 01/05/2004] [Revised: 02/06/2004] [Accepted: 02/23/2004] [Indexed: 11/28/2022]
Abstract
The clinical response to antitumor therapy is measured using imaging, such as CT or MRI, 6-12 weeks following chemotherapy treatment. The images at that time reflect both tumor cell death and new growth. Therefore, the amount of tumor cell death caused by chemotherapy cannot be efficiently quantified with current imaging modalities. A quantitative measurement of tumor cell death immediately following chemotherapy is needed to help validate both new agents and to optimize administration of existing therapies. Annexin V is a 36kD protein that binds to exposed phosphatidylserine (PS) on dying cells. In order to synthesize a probe that can detect cell death in vivo, the positron emitter F-18 was conjugated to annexin V via the compound N- succinimidyl-4-[18F]fluorobenzoate, [18F]SFB. The decay corrected radiochemical yield of F-18 labeled annexin V from 18F fluoride was 17.6 +/- 5.6% (n = 4) in three hours. The stepwise radiochemical yield of the conjugation step with annexin V was as high as 70% when a protein concentration of 5 mg/ml was used. Cancer cells treated with the chemotherapeutic agent, etoposide, showed an 88% increase in the binding of F-18 labeled annexin V compared to untreated cells. We conclude that [18F] labeled annexin V can be readily prepared by the conjugation of annexin V with [18F]SFB and that the positron-emitting compound is biologically active in detecting apoptosis.
Collapse
Affiliation(s)
- J Toretsky
- Georgetown University, Washington, DC 20007, USA.
| | | | | | | | | | | |
Collapse
|
14
|
Kartachova M, Haas RLM, Olmos RAV, Hoebers FJP, van Zandwijk N, Verheij M. In vivo imaging of apoptosis by 99mTc-Annexin V scintigraphy: visual analysis in relation to treatment response. Radiother Oncol 2005; 72:333-9. [PMID: 15450733 DOI: 10.1016/j.radonc.2004.07.008] [Citation(s) in RCA: 91] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 05/27/2004] [Revised: 06/26/2004] [Accepted: 06/28/2004] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Anticancer therapy induces apoptosis in a dose- and time-dependent fashion. (99m)Tc-Hynic-rh-Annexin V scintigraphy (TAVS) enables non-invasive in vivo imaging of treatment-induced apoptosis. We identified the visual patterns of (99m)Tc-Hynic-rh-Annexin V tumour uptake and related these to treatment response. PATIENTS AND METHODS Thirty-three patients with malignant lymphoma (n=26), leukaemia (n=1) NSCLC (n=5), H&NSCC (n=1), scheduled for radiotherapy (n=27), platinum-based chemotherapy (n=5) or concurrent chemoradiation (n=1), underwent TAVS before and early after the start of treatment. Planar and SPECT images were visually examined to assess changes in tumour (99m)Tc-Hynic-rh-Annexin V uptake. Twenty-nine patients were eligible for further analysis. Annexin V uptake before (U(baseline)) and early after (U(post)) the start of treatment was graded using a four-step scale: 0, absent; 1, weak; 2, moderate and 3, intense. The difference between these values (Delta U) was calculated and correlated to tumour response after therapy (Spearman rank correlation test). RESULTS Weak to moderate U(baseline) was detected in 13/15 patients with a complete response and U(post) was markedly increased in all these cases (Delta U range 1-3). Partial response (n=7) was associated with weak to moderate U(baseline) and a moderately increased U(post) (Delta U range 1-2). In patients with stable disease (n=5), U(baseline) was predominantly weak, without considerable changes in uptake after the start of treatment (Delta U range 0-1). Finally, in case of progressive disease (n=2), either no tumour uptake or a decrease in U(post) was detected (Delta U=-1). A statistically significant correlation was found between changes in (99m)Tc-Hynic-rh-Annexin V tumour uptake and clinical response (correlation coefficient=0.62; P<0.001). CONCLUSIONS Complete or partial tumour response was associated with a marked increase of (99m)Tc Hynic-rh-Annexin V accumulation early during treatment compared to baseline values. In case of stable or progressive disease, pretreatment scans demonstrated predominantly low (99m)Tc Hynic-rh-Annexin V tumour uptake and no significant increase early after treatment. These results indicate that TAVS might be useful as a predictive test for treatment response.
Collapse
Affiliation(s)
- Marina Kartachova
- Department of Nuclear Medicine, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | | | | | | | | | | |
Collapse
|
15
|
Belhocine TZ, Tait JF, Vanderheyden JL, Li C, Blankenberg FG. Nuclear medicine in the era of genomics and proteomics: lessons from annexin V. J Proteome Res 2004; 3:345-9. [PMID: 15253415 DOI: 10.1021/pr049968a] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/22/2023]
Abstract
In the past decade, genomics and proteomics have begun to develop many new targets for potential diagnostic and therapeutic agents. Among the life sciences, nuclear medicine is also deeply involved in the field of clinical investigation. Experience with radiolabeled annexin V highlights the many steps required to translate a good basic-science concept into the clinical setting. This model also emphasizes the value of synergy between basic and medical specialties in developing and optimizing a clinically useful product initially derived from basic investigation.
Collapse
Affiliation(s)
- Tarik Z Belhocine
- Department of Nuclear Medicine, Jules Bordet Cancer Institute, Brussels, Belgium.
| | | | | | | | | |
Collapse
|
16
|
Haas RLM, de Jong D, Valdés Olmos RA, Hoefnagel CA, van den Heuvel I, Zerp SF, Bartelink H, Verheij M. In vivo imaging of radiation-induced apoptosis in follicular lymphoma patients. Int J Radiat Oncol Biol Phys 2004; 59:782-7. [PMID: 15183481 DOI: 10.1016/j.ijrobp.2003.11.017] [Citation(s) in RCA: 96] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Received: 08/04/2003] [Revised: 11/11/2003] [Accepted: 11/13/2003] [Indexed: 11/17/2022]
Abstract
PURPOSE To evaluate (99m)Tc-Annexin-V (TAV) scintigraphy in monitoring radiation-induced apoptotic cell death in follicular lymphoma (FL) patients. PATIENTS AND METHODS Eleven FL patients (7 female and 4 male; median age, 58 years; range, 42-80 years) with recurrent disease underwent TAV imaging before and 24 hours after the last fraction of the 2 x 2 Gy involved field radiotherapy regimen. Fine-needle aspiration cytology was performed on 5 consecutive days to determine the optimal time window for apoptosis detection and to confirm the apoptotic nature of the response. The TAV scintigraphy (total body studies and SPECT of the irradiated sites) was performed 4 hours after the administration of the radiopharmaceutical. Tumor uptake was scored in a semiquantitative manner as absent (-) weak (+/-), present (+), or intense (++) with corresponding categories for the cytologic slides. Response evaluation was performed after 1 week and 4 weeks both in terms of completeness and speed of remission. RESULTS Baseline TAV uptake was absent in 6 and weak in 5 patients. Sequential cytology indicated that the optimal time period for apoptosis assessment was between 24 and 48 hours after the last fraction of the 2 x 2 Gy regimen. Baseline cytology was concordant with baseline TAV in all patients. Apoptotic feature appearance (nuclear chromatin condensation, margination and apoptotic body formation) after low-dose irradiation matched the irradiation response in all patients. In all but 1 patient the posttreatment TAV uptake matched the posttreatment cytology. In these 10 patients the cytology and TAV results correlated with the type and onset of the clinical response. CONCLUSION Tumor (99m)Tc-Annexin-V uptake can be increased after 2 x 2 Gy involved field radiotherapy. This increase was concordant with the appearance of apoptotic morphology as determined by cytology, and correlated with the clinical outcome. Apoptotic cell death can be observed on Day 4 of this regimen and if so predicts a complete remission within 1 week.
Collapse
Affiliation(s)
- Rick L M Haas
- Department of Radiotherapy, The Netherlands Cancer Institute/Antoni van Leeuwenhoek Hospital, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | |
Collapse
|
17
|
Abstract
Molecular imaging aims to visualize the cellular and molecular processes occurring in living tissues, and for the imaging of specific molecules in vivo, the development of reporter probes and dedicated imaging equipment is most important. Reporter genes can be used to monitor the delivery and magnitude of therapeutic gene transfer, and the time variation involved. Imaging technologies such as micro-PET, SPECT, MRI and CT, as well as optical imaging systems, are able to non-invasively detect, measure, and report the simultaneous expression of multiple meaningful genes. It is believed that recent advances in reporter probes, imaging technologies and gene transfer strategies will enhance the effectiveness of gene therapy trials.
Collapse
Affiliation(s)
- E Edmund Kim
- Department of Radiology and Medicine, Center for Metabolic Imaging, Nuclear Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
18
|
Lahorte CMM, Vanderheyden JL, Steinmetz N, Van de Wiele C, Dierckx RA, Slegers G. Apoptosis-detecting radioligands: current state of the art and future perspectives. Eur J Nucl Med Mol Imaging 2004; 31:887-919. [PMID: 15138718 DOI: 10.1007/s00259-004-1555-4] [Citation(s) in RCA: 128] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/15/2022]
Abstract
This review provides a critical and thorough overview of the radiopharmaceutical development and in vivo evaluation of all apoptosis-detecting radioligands that have emerged so far, along with their possible applications in nuclear medicine. The following SPECT and PET radioligands are discussed: all forms of halogenated Annexin V (i.e. (123)I-labelled, (124)I-labelled, (125)I-labelled, (18)F-labelled), (99m)Tc/(94m)Tc-labelled Annexin V derivatives using different chelators and co-ligands (i.e. BTAP, Hynic, iminothiolane, MAG(3), EDDA, EC, tricarbonyl, SDH) or direct (99m)Tc-labelling, (99m)Tc-labelled Annexin V mutants and (99m)Tc/(18)F-radiopeptide constructs (i.e. AFIM molecules), (111)In-DTPA-PEG-Annexin V, (11)C-Annexin V and (64)Cu-, (67)Ga- and (68)Ga-DOTA-Annexin V. In addition, the potential role and clinical relevance of anti-PS monoclonal antibodies and other alternative apoptosis markers are reviewed, including: anti-Annexin V monoclonal antibodies, radiolabelled caspase inhibitors and substrates and mitochondrial membrane permeability targeting radioligands. Nevertheless, major emphasis is placed on the group of Annexin V-based radioligands, in particular (99m)Tc-Hynic-Annexin V, since this molecule is by far the most extensively investigated and best-characterised apoptosis marker at present. Furthermore, the newly emerging imaging modalities for in vivo detection of programmed cell death, such as MRI, MRS, optical, bioluminescent and ultrasound imaging, are briefly described. Finally, some future perspectives are presented with the aim of promoting the development of potential new strategies in pursuit of the ideal cell death-detecting radioligand.
Collapse
Affiliation(s)
- Christophe M M Lahorte
- Department of Radiopharmacy, Faculty of Pharmaceutical Sciences, Ghent University, Harelbekestraat 72, 9000 Ghent, Belgium.
| | | | | | | | | | | |
Collapse
|
19
|
Collingridge DR, Glaser M, Osman S, Barthel H, Hutchinson OC, Luthra SK, Brady F, Bouchier-Hayes L, Martin SJ, Workman P, Price P, Aboagye EO. In vitro selectivity, in vivo biodistribution and tumour uptake of annexin V radiolabelled with a positron emitting radioisotope. Br J Cancer 2003; 89:1327-33. [PMID: 14520468 PMCID: PMC2394302 DOI: 10.1038/sj.bjc.6601262] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 12/20/2022] Open
Abstract
The availability of a noninvasive method to detect and quantify apoptosis in tumours will enable tumour response to several cancer therapies to be assessed. We have synthesised two radiotracers, annexin V and the N-succinimidyl-3-iodobenzoic acid (SIB) derivative of annexin V, labelled with radio-iodine (124I and 125I) and provided proof of the concept by assessing specific binding and biodistribution of these probes to apoptotic cells and tumours. We have also assessed the tumour uptake of [124I]annexin V in a mouse model of apoptosis. RIF-1 cells induced to undergo apoptosis in vitro showed a drug concentration-dependent increased binding of [125I]annexin V and [125I]SIB–annexin V. In the same model system, there was an increase in terminal deoxynucleotidyl transferase-mediated nick end labelling (TUNEL)-positive cells and a decrease in clonogenic survival. Radiotracer binding was completely inhibited by preincubation with unlabelled annexin V. In RIF-1 tumour-bearing mice, rapid distribution of [125I]SIB–annexin V-derived radioactivity to kidneys was observed and the radiotracer accumulated in urine. The binding of [125I]SIB–annexin V to RIF-1 tumours increased by 2.3-fold at 48 h after a single intraperitoneal injection of 5-fluorouracil (165 mg kg−1 body weight), compared to a 4.4-fold increase in TUNEL-positive cells measured by immunostaining. Positron emission tomography images with both radiotracers demonstrated intense localisation in the kidneys and bladder. Unlike [124I]SIB–annexin V, [124I]annexin V also showed localisation in the thyroid region presumably due to deiodination of the radiolabel. [124I]SIB–annexin V is an attractive candidate for in vivo imaging of apoptosis by PET.
Collapse
Affiliation(s)
- D R Collingridge
- Cancer Research UK PET Oncology Group, Department of Cancer Medicine, Imperial College of Science Technology and Medicine, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - M Glaser
- Imaging Research Solutions Limited, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - S Osman
- Imaging Research Solutions Limited, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - H Barthel
- Cancer Research UK PET Oncology Group, Department of Cancer Medicine, Imperial College of Science Technology and Medicine, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Department of Nuclear Medicine, University of Leipzig, Liebigstrasse 20A, 04103 Leipzig, Germany
| | - O C Hutchinson
- Cancer Research UK PET Oncology Group, Department of Cancer Medicine, Imperial College of Science Technology and Medicine, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - S K Luthra
- Imaging Research Solutions Limited, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - F Brady
- Imaging Research Solutions Limited, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - L Bouchier-Hayes
- The Smurfit Institute of Genetics, Trinity College, Dublin, Ireland
| | - S J Martin
- The Smurfit Institute of Genetics, Trinity College, Dublin, Ireland
| | - P Workman
- Cancer Research UK Centre for Cancer Therapeutics, Institute of Cancer Research, Sutton, Surrey SM2 SNG, UK
| | - P Price
- Cancer Research UK PET Oncology Group, Department of Cancer Medicine, Imperial College of Science Technology and Medicine, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
| | - E O Aboagye
- Cancer Research UK PET Oncology Group, Department of Cancer Medicine, Imperial College of Science Technology and Medicine, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK
- Cancer Research UK PET Oncology Group, Department of Cancer Medicine, Imperial College of Science Technology and Medicine, MRC Cyclotron Building, Hammersmith Hospital, Du Cane Road, London W12 0NN, UK. E-mail:
| |
Collapse
|
20
|
Sarwal M, Chua MS, Kambham N, Hsieh SC, Satterwhite T, Masek M, Salvatierra O. Molecular heterogeneity in acute renal allograft rejection identified by DNA microarray profiling. N Engl J Med 2003; 349:125-38. [PMID: 12853585 DOI: 10.1056/nejmoa035588] [Citation(s) in RCA: 540] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Academic Contribution Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND The causes and clinical course of acute rejection vary, and it is not possible to predict graft outcome reliably on the basis of available clinical, pathological, and genetic markers. We hypothesized that previously unrecognized molecular heterogeneity might underlie some of the variability in the clinical course of acute renal allograft rejection and in its response to treatment. METHODS We used DNA microarrays in a systematic study of gene-expression patterns in biopsy samples from normal and dysfunctional renal allografts. A combination of exploratory and supervised bioinformatic methods was used to analyze these profiles. RESULTS We found consistent differences among the gene-expression patterns associated with acute rejection, nephrotoxic effects of drugs, chronic allograft nephropathy, and normal kidneys. The gene-expression patterns associated with acute rejection suggested at least three possible distinct subtypes of acute rejection that, although indistinguishable by light microscopy, were marked by differences in immune activation and cellular proliferation. Since the gene-expression patterns pointed to substantial variation in the composition of immune infiltrates, we used immunohistochemical staining to define these subtypes further. This analysis revealed a striking association between dense CD20+ B-cell infiltrates and both clinical glucocorticoid resistance (P=0.01) and graft loss (P<0.001). CONCLUSIONS Systematic analysis of gene-expression patterns provides a window on the biology and pathogenesis of renal allograft rejection. Biopsy samples from patients with acute rejection that are indistinguishable on conventional histologic analysis reveal extensive differences in gene expression, which are associated with differences in immunologic and cellular features and clinical course. The presence of dense clusters of B cells in a biopsy sample was strongly associated with severe graft rejection, suggesting a pivotal role of infiltrating B cells in acute rejection.
Collapse
Affiliation(s)
- Minnie Sarwal
- Department of Pediatrics, Stanford University, Stanford, Calif, USA.
| | | | | | | | | | | | | |
Collapse
|
21
|
Affiliation(s)
- Ismet Sarikaya
- Section of Nuclear Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA.
| | | | | | | |
Collapse
|
22
|
Abstract
Myocardial necrosis plays an important role in the pathogenesis of various cardiovascular disorders and can result from different myocardial insults. Its non-invasive identification and localisation therefore may help in the diagnosis of these disorders, as well as in prognosis and assessment of treatment response. Apoptosis, or programmed cell death, is important in the spectrum of myocardial damage since it is gradually becoming more apparent that cell death may begin as apoptosis and not as necrosis. First attempts to directly visualise the area of myocardial necrosis were based on recognition of myocardial infarction with "hot spot imaging agents" in patients with chest pain. Since then, the study of myocardial necrosis with gamma imaging agents has gone beyond the detection of myocardial infarction, and attempts have been made to diagnose other cardiovascular disorders associated with cardiac cell death such as heart transplant rejection, myocarditis, cardiotoxicity and cardiomyopathies. Traditionally, two hot spot imaging agents have been used for the detection of myocardial necrosis, (99m)Tc-pyrophosphate and (111)In-antimyosin. In addition, preliminary studies have demonstrated promising results with (99m)Tc-glucarate. Recently, (99m)Tc-annexin V has been successfully used for non-invasive gamma imaging of apoptosis after acute myocardial infarction, acute myocardial ischaemia, acute cardiac allograft rejection and malignant intracardiac tumours. This review article focusses on the characteristics of these different myocardial necrotic and apoptotic markers and compares their role in the assessment of myocardial damage.
Collapse
|
23
|
Abstract
The biochemical and physiological processes involved in apoptosis were described from the perspective of detection by standard, clinical, noninvasive imaging modalities. The difficulties of monitoring apoptosis in vivo were discussed. Magnetic resonance imaging (MRI) approaches used to study apoptosis were surveyed. The cell shrinkage associated with apoptosis can be detected due to changes in tissue water T(2) and T(1)rho relaxation times and apparent diffusion coefficient (ADC). Magnetic resonance spectroscopy (MRS) approaches used to study apoptosis in vivo have largely centered on the formation of cytoplasmic lipid bodies, detected by 1H MRS, and metabolic/bioenergetic changes detected by 31P and 13C MRS. The most successful approach to in vivo mapping of apoptosis uses the high specific binding of annexin V or synaptotagmin I to phosphatidylserine (PS) that appears on the extracellular plasma membrane of cells during apoptosis. Technetium-99m (99mTc)-radiolabeling of the annexin V and superparamagnetic iron oxide (SPIO) labeling of the C2 domain of synaptotagmin I allow good in vivo apoptosis detection by gamma camera imaging and MRI, respectively.
Collapse
Affiliation(s)
- Manfred Brauer
- Department of Chemistry and Biochemistry, University of Guelph, N1G 2W1, Guelph, Ontario, Canada.
| |
Collapse
|
24
|
Signore A, Annovazzi A, Corsetti F, Capriotti G, Chianelli M, De Winter F, Scopinaro F. Biological imaging for the diagnosis of inflammatory conditions. BioDrugs 2003; 16:241-59. [PMID: 12196038 DOI: 10.2165/00063030-200216040-00002] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 01/07/2023]
Abstract
Radiopharmaceuticals used for in vivo imaging of inflammatory conditions can be conveniently classified into six categories according to the different phases in which the inflammatory process develops. The trigger of an inflammatory process is a pathogenic insult (phase I) that causes activation of endothelial cells (phase II); there is then an increase of vascular permeability followed by tissue oedema (phase III). Phase IV is characterised by infiltration of polymorphonuclear cells, and a self-limiting regulatory process called apoptosis is observed (phase V). If the inflammatory process persists, late chronic inflammation takes place (phase VI). In some pathological conditions, such as organ-specific autoimmune diseases, chronic inflammation is present early in the disease. The aim of nuclear medicine in the field of inflammation/infection is to develop noninvasive tools for the in vivo detection of specific cells and tissues. This would allow early diagnosis of initial pathophysiological changes that are undetectable by clinical examination or by other diagnostic tools, and could also be used to evaluate the state of activity of the disease during therapy. These potential applications are of great interest in clinical practice. In this review, we describe the various approaches that have been developed in the last 25 years of experience. Recent advances in the diagnosis of inflammatory processes have led to the development of specific radiopharmaceuticals that are intended to allow specific stage-related diagnosis.
Collapse
Affiliation(s)
- Alberto Signore
- Department of Clinical Sciences, Nuclear Medicine, 2nd Faculty of Medicine, University of Rome, La Sapienza, Rome, Italy.
| | | | | | | | | | | | | |
Collapse
|
25
|
Sharma V, Luker GD, Piwnica-Worms D. Molecular imaging of gene expression and protein function in vivo with PET and SPECT. J Magn Reson Imaging 2002; 16:336-51. [PMID: 12353250 DOI: 10.1002/jmri.10182] [Citation(s) in RCA: 92] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/10/2022] Open
Abstract
Molecular imaging is broadly defined as the characterization and measurement of biological processes in living animals, model systems, and humans at the cellular and molecular level using remote imaging detectors. One underlying premise of molecular imaging is that this emerging field is not defined by the imaging technologies that underpin acquisition of the final image per se, but rather is driven by the underlying biological questions. In practice, the choice of imaging modality and probe is usually reduced to choosing between high spatial resolution and high sensitivity to address a given biological system. Positron emission tomography (PET) and single-photon emission computed tomography (SPECT) inherently use image-enhancing agents (radiopharmaceuticals) that are synthesized at sufficiently high specific activity to enable use of tracer concentrations of the compound (picomolar to nanomolar) for detecting molecular signals while providing the desired levels of image contrast. The tracer technologies strategically provide high sensitivity for imaging small-capacity molecular systems in vivo (receptors, enzymes, transporters) at a cost of lower spatial resolution than other technologies. We review several significant PET and SPECT advances in imaging receptors (somatostatin receptor subtypes, neurotensin receptor subtypes, alpha(v)beta(3) integrin), enzymes (hexokinase, thymidine kinase), transporters (MDR1 P-glycoprotein, sodium-iodide symporter), and permeation peptides (human immunodeficiency virus type 1 (HIV-1) Tat conjugates), as well as innovative reporter gene constructs (herpes simplex virus 1 thymidine kinase, somatostatin receptor subtype 2, cytosine deaminase) for imaging gene promoter activation and repression, signal transduction pathways, and protein-protein interactions in vivo.
Collapse
Affiliation(s)
- Vijay Sharma
- Molecular Imaging Center, Mallinckrodt Institute of Radiology and Department of Molecular Biology and Pharmacology, Washington University Medical School, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
26
|
Abstract
The requirement for cytotoxic T lymphocytes during allograft rejection is controversial. We have demonstrated that CD8+ T cells are not essential for allograft rejection or for the induction of apoptosis in two experimental models of transplantation. To determine candidate cells types which may play a role in the events leading to graft rejection, the cellular composition of rejecting allografts was determined. We demonstrate that substantial numbers of NK cells, of recipient origin, infiltrate allografts as early as 12 h after transplantation. These NK cells produce cytokines and express cytotoxic mediators such as granzyme B and FasL. It is unknown which NK cell receptors are expressed and activated during transplantation. NK cells express multiple cell surface receptors, including MHC class I binding inhibitory receptors, which deliver a negative signal, and activation receptors, which stimulate cytokine secretion and cytotoxicity of NK cells. To begin to understand NK cell activation in the context of transplantation, we have recently cloned a novel rat immunoglobulin-like surface receptor from a rejecting liver allograft. Sequence analysis demonstrates that this putative activation receptor contains 71% identity to human NKp30 at the DNA level, suggesting that it is the rat homologue (rNKp30). Characterization of NK activation receptors may lead to better understanding of the interactions between the innate and adaptive immune responses in transplantation.
Collapse
Affiliation(s)
- Christine L Hsieh
- Department of Surgery, Stanford University School of Medicine, CA 94305-5492, USA
| | | | | | | | | |
Collapse
|
27
|
Ogura Y, Martinez OM, Villanueva JC, Tait JF, Strauss HW, Higgins JP, Tanaka K, Esquivel CO, Blankenberg FG, Krams SM. Apoptosis and allograft rejection in the absence of CD8+ T cells. Transplantation 2001; 71:1827-34. [PMID: 11455265 DOI: 10.1097/00007890-200106270-00020] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND The requirement for cytotoxic T lymphocytes during allograft rejection is controversial. We previously demonstrated that CD8+ T cells are not necessary for allograft rejection or for the induction of apoptosis in rat small intestinal transplantation. In this study, we examined the mechanisms of apoptosis and rejection after liver transplantation in the absence of CD8+ T cells. METHODS Either Lewis or dark agouti rat liver grafts were transplanted into Lewis recipients to create syngeneic and allogeneic combinations. CD8+ T cells were depleted in an additional allogeneic group by treatment with OX-8 mAb on day -1 and day 1 after liver transplant. RESULTS Apoptosis and rejection were observed in both the CD8+ T cell-depleted allogeneic and allogeneic grafts by hematoxylin and eosin staining, terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining, and radiolabeled-annexin V in vivo imaging. Granzyme B and FasL were expressed in all allogeneic transplants, including those depleted of CD8+ T cells, indicating that a mononuclear cell other than a CD8+ T cell can be the source of these molecules during allograft rejection. Activation of the caspase cascade was detected in all rejecting allografts. Caspases 3, 8, and 9 were activated at similar significantly elevated levels in both allogeneic and CD8+ T cell-depleted liver grafts. CONCLUSION These data indicate that in the absence of CD8+ T cells an alternative pathway, associated with granzyme B and FasL expression and activation of the caspase cascade, can mediate apoptosis and graft rejection.
Collapse
Affiliation(s)
- Y Ogura
- Transplant Immunobiology Laboratory, Department of Surgery, Stanford University School of Medicine, 1201 Welch Road, Stanford, CA 94305-5492, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
The term molecular imaging can be broadly defined as the in vivo characterization and measurement of biologic processes at the cellular and molecular level. In contradistinction to "classical" diagnostic imaging, it sets forth to probe the molecular abnormalities that are the basis of disease rather than to image the end effects of these molecular alterations. While the underlying biology represents a new arena for many radiologists, concomitant efforts such as development of novel agents, signal amplification strategies, and imaging technologies clearly dovetail with prior research efforts of our specialty. Radiologists will play a leading role in directing developments of this embryonic but burgeoning field. This article presents some recent developments in molecular sciences and medicine and shows how imaging can be used, at least experimentally, to assess specific molecular targets. In the future, specific imaging of such targets will allow earlier detection and characterization of disease, earlier and direct molecular assessment of treatment effects, and a more fundamental understanding of the disease process.
Collapse
Affiliation(s)
- R Weissleder
- Center for Molecular Imaging Research, Department of Radiology, Massachusetts General Hospital, Harvard Medical School, Bldg 149, Rm 5403, Charlestown, MA 02129, USA. weissler\
| | | |
Collapse
|
29
|
Yang DJ, Azhdarinia A, Wu P, Yu DF, Tansey W, Kalimi SK, Kim EE, Podoloff DA. In vivo and in vitro measurement of apoptosis in breast cancer cells using 99mTc-EC-annexin V. Cancer Biother Radiopharm 2001; 16:73-83. [PMID: 11279800 DOI: 10.1089/108497801750096087] [Citation(s) in RCA: 54] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/12/2022] Open
Abstract
OBJECTIVE The purpose of this study was to develop an imaging technique to measure and monitor tumor cells undergoing programmed death caused by radiation and chemotherapy using 99mTc-EC-annexin V. Annexin V has been used to measure programmed cell death both in vitro and in vivo. Assessment of apoptosis would be useful to evaluate the efficacy and mechanisms of therapy and disease progression or regression. METHODS Ethylenedicysteine (EC) was conjugated to annexin V using sulfo-N-hydroxysuccinimide and 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide-HCl as coupling agents. The yield of EC-annexin V was 100%. In vitro cellular uptake, pre- and post-radiation (10-30 Gy) and paclitaxel treatment, was quantified using 99mTc-EC-annexin V. Tissue distribution and planar imaging of 99mTc-EC-annexin V were determined in breast tumor-bearing rats at 0.5, 2, and 4 hrs. To demonstrate in vivo cell apoptosis that occurred during chemotherapy, a group of rats was treated with paclitaxel and planar imaging studies were conducted at 0.5-4 hrs. Computer outlined region of interest (ROI) was used to quantify tumor uptake on day 3 and day 5 post-treatment. RESULTS In vitro cellular uptake showed that there was significantly increased uptake of 99mTc-EC-annexin V after irradiation (10-30 Gy) and paclitaxel treatment. In vivo biodistribution of 99mTc-EC-annexin in breast tumor-bearing rats showed increased tumor-to-blood, tumor-to-lung and tumor-to-muscle count density ratios as a function of time. Conversely, tumor-to-blood count density ratios showed a time-dependent decrease with 99mTc-EC in the same time period. Planar images confirmed that the tumors could be visualized clearly with 99mTc-EC-annexin. There was a significant difference of ROI ratios between pre- and post-paclitaxel treatment groups at 2 and 4 hrs post injection. CONCLUSION The results indicate that apoptosis can be quantified using 99mTc-EC-annexin and that it is feasible to use 99mTc-EC-annexin to image tumor apoptosis.
Collapse
Affiliation(s)
- D J Yang
- Univ. of Texas M. D. Anderson Cancer Center, Dept. of Nuclear Medicine, Box 59, 1515 Holcombe Boulevard, Houston, Texas 77030, USA.
| | | | | | | | | | | | | | | |
Collapse
|
30
|
Dry GM, Yasinskaya YI, Williams JK, Ehrlich GD, Preston RA, Hu FZ, Gruss JS, Ellenbogen RG, Cunningham ML. Inhibition of apoptosis: a potential mechanism for syndromic craniosynostosis. Plast Reconstr Surg 2001; 107:425-32. [PMID: 11214058 DOI: 10.1097/00006534-200102000-00019] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/26/2022]
Abstract
The biologic pathogenesis of syndromic craniosynostosis remains unknown. The purpose of this investigation was to determine whether specific biologic differences exist between normal calvarial osteoblasts and osteoblasts derived from patients with syndromic craniosynostosis. This study (1) examined the apoptotic rate and cell cycle of osteoblasts derived from patients with syndromic craniosynostosis, and (2) investigated for the presence of soluble factors released from syndrome-derived osteoblasts. Osteoblast cell lines were established from calvarial specimens of patients with clinically diagnosed syndromic synostosis and from normal controls. A co-culture technique was used to investigate for the presence of elaborated soluble factors. Apoptotic rate and cell cycle analyses were performed by using flow cytometry after staining with annexin V-fluorescein isothiocyanate and propidiumiodide, respectively. The apoptotic rate was significantly reduced in syndrome-derived osteoblasts as compared with control osteoblasts. Control osteoblasts co-cultured with syndromic osteoblasts demonstrated a dramatic reduction in their apoptotic rate as compared with those co-cultured with control osteoblasts. These results indicate that osteoblasts derived from patients with syndromic craniosynostosis display a lower apoptotic rate, a normal DNA synthetic rate, and the capability to reduce the apoptotic rate in normal calvarial osteoblasts through the elaboration of soluble factors.
Collapse
Affiliation(s)
- G M Dry
- Children's Craniofacial Center, Children's Hospital, University of Washington, Seattle 98195-6320, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
31
|
D'Arceuil H, Rhine W, de Crespigny A, Yenari M, Tait JF, Strauss WH, Engelhorn T, Kastrup A, Moseley M, Blankenberg FG. 99mTc annexin V imaging of neonatal hypoxic brain injury. Stroke 2000; 31:2692-700. [PMID: 11062296 DOI: 10.1161/01.str.31.11.2692] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Academic Contribution Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Delayed cell loss in neonates after cerebral hypoxic-ischemic injury (HII) is believed to be a major cause of cerebral palsy. In this study, we used radiolabeled annexin V, a marker of delayed cell loss (apoptosis), to image neonatal rabbits suffering from HII. METHODS Twenty-two neonatal New Zealand White rabbits had ligation of the right common carotid artery with reduction of inspired oxygen concentration to induce HII. Experimental animals (n=17) were exposed to hypoxia until an ipsilateral hemispheric decrease in the average diffusion coefficient occurred. After reversal of hypoxia and normalization of average diffusion coefficient values, experimental animals were injected with (99m)Tc annexin V. Radionuclide images were recorded 2 hours later. RESULTS Experimental animals showed no MR evidence of blood-brain barrier breakdown or perfusion abnormalities after hypoxia. Annexin images demonstrated multifocal brain uptake in both hemispheres of experimental but not control animals. Histology of the brains from experimental animals demonstrated scattered pyknotic cortical and hippocampal neurons with cytoplasmic vacuolization of glial cells without evidence of apoptotic nuclei by terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) staining. Double staining with markers of cell type and exogenous annexin V revealed that annexin V was localized in the cytoplasm of scattered neurons and astrocytes in experimental and, less commonly, control brains in the presence of an intact blood-brain barrier. CONCLUSIONS Apoptosis may develop after HII even in brains that appear normal on diffusion-weighted and perfusion MR. These data suggest a role of radiolabeled annexin V screening of neonates at risk for the development of cerebral palsy.
Collapse
Affiliation(s)
- H D'Arceuil
- Department of Radiology, Stanford University School of Medicine, Stanford, CA 94305-5105, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|