4.6

CiteScore

2.2

Impact Factor
  • ISSN 1674-8301
  • CN 32-1810/R
Haoran Jia, Tianwu Xie. Tracers progress for positron emission tomography imaging of glial-related disease[J]. The Journal of Biomedical Research, 2022, 36(5): 321-335. DOI: 10.7555/JBR.36.20220017
Citation: Haoran Jia, Tianwu Xie. Tracers progress for positron emission tomography imaging of glial-related disease[J]. The Journal of Biomedical Research, 2022, 36(5): 321-335. DOI: 10.7555/JBR.36.20220017

Tracers progress for positron emission tomography imaging of glial-related disease

Funds: This work was supported by the Key Laboratory of Biomedical Engineering of Hainan Province under Opening Foundation 2022001.
More Information
  • Corresponding author:

    Tianwu Xie, Institute of Radiation Medicine, Fudan University, 2094 Xietu Road, Shanghai 200032, China. Tel: +86-21-64048363, E-mail: tianwuxie@fudan.edu.cn

  • Received Date: January 22, 2022
  • Revised Date: April 22, 2022
  • Accepted Date: May 23, 2022
  • Available Online: June 27, 2022
  • Glial cells play an essential part in the neuron system. They can not only serve as structural blocks in the human brain but also participate in many biological processes. Extensive studies have shown that astrocytes and microglia play an important role in neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, Huntington's disease, as well as glioma, epilepsy, ischemic stroke, and infections. Positron emission tomography is a functional imaging technique providing molecular-level information before anatomic changes are visible and has been widely used in many above-mentioned diseases. In this review, we focus on the positron emission tomography tracers used in pathologies related to glial cells, such as glioma, Alzheimer's disease, and neuroinflammation.
  • The brain serves as the center of the nervous system composed primarily of two broad classes of cells: neurons and glial cells. Neurons send signals to specific target cells over long distances through axons. Most of the space in the brain is taken up by axons, which are often bundled together as nerve fiber tracts. Some axons are wrapped in a fatty insulating sheath of white myelin, making parts of the brain filled exclusively with nerve fibers appear as light-colored white matter, in contrast to the darker-colored grey matter that marks areas with high densities of neuron cell bodies. Although neurons are usually considered as the most important cells in the brain, glial cells, which are abundant in white matter, also play an essential part in the central nerve system (CNS).

    Glial cells, also called glia or neuroglia, were originally believed to just provide structural support, and had a glia-neuron ratio (GNR) of 10:1, which are both proven wrong nowadays. It's now believed that GNRs vary in different regions of the brain and have an average number of less than 1:1[1]. Therefore, with more diversities than neurons, glial cells take part in 33% to 66% of the total brain mass[2]. In the CNS, glial cells consist of oligodendrocytes, astrocytes, ependymal cells, and microglia. They have four main functions: (1) to surround neurons and hold them in place; (2) to supply nutrients and oxygen to neurons; (3) to insulate one neuron from another; (4) to destroy pathogens and remove damaged or degenerative neurons.

    Because of the heterogeneity of glial cells population, there are wide ranges of different neuro-pathologies and non-neural diseases related to glial disorders. Astrocytes and microglia play an important role in neurodegenerative disease, such as Alzheimer's disease (AD), Parkinson's disease (PD) and Huntington disease, as well as glioma, epilepsy, ischemic stroke and many kinds of infections[35].

    Neuroinflammation is an inflammatory response in the CNS, typically caused by cell damaging processes such as infection, traumatic brain injury, toxic metabolites and many other cues. During neuroinflammation, CNS immune glial cells will be activated and produce factors that mediate neuroinflammation process. There are both positive and negative aspects of neuroinflammation, depending on its intensity and duration[6].

    In cellular level, the immune activation in the brain is related to microglia and astrocytes, which generally aimed at evoking CNS immunity of removing harmful agents, repairing damaged tissue and maintaining homeostasis in non-pathological conditions. Under diseased conditions, both microglia and astrocytes would present phenotype changes from resting morphology to reactive phenotype, with function alternation and affecting the inflammatory process[78]. Chronic or exaggerated microglial activation may cause damage in the CNS and takes an important role in the process of synaptic dysfunction, neurogenesis inhibition, and neurodegenerative diseases[9], such as AD, PD, amyotrophic lateral sclerosis, and frontotemporal dementia. In those diseases, the immune system may deactivate and even worse, neuroinflammation may contribute to disease processes. Inflammatory and neoplastic brain disorders also relate to neuroinflammatory reactions.

    Glioma is another type of disease related to glial cells and is one of the most popular primary intracranial tumors which constitutes about 60% of all cerebral tumors[10]. According to the World Health Organization (WHO) classification of central nervous system tumors, gliomas can be divided into astrocytomas, oligodendrogliomas, and glioblastomas[11]. Although the cell origin of gliomas remains elusive, it may be closely related to all kinds of glial cells and glial progenitor cells[12].

    Positron emission tomography (PET) is a functional imaging technique capable of detecting biochemical processes as well as the expression of some proteins. It provides molecular-level information before anatomic changes are visible, while computed tomography (CT) and magnetic resonance imaging (MRI) scans can only show images in the level of organs and tissues. PET imaging depends on radiolabelled molecular probes with different rates of uptake in various tissues. Therefore, selection of PET tracers is one of the key points for obtaining precise and meaningful diagnostic message. With PET imaging, glial phenotype can be distinguished in vivo. In this article, we summarize the tracers targeted to glial cells for PET imaging.

    Neuroinflammation is highly related to glial cells of inactive microglia and astrocytes with different phenotypes, where most of the imaging targets for neuroinflammation are located.

    The first and most used neuroinflammation biomarker is translocator protein 18 kDa (TSPO, historically known as peripheral-type benzodiazepine receptor, PBR). TSPO is responsible for the translocation of cholesterol[13], and involved in many physiological functions such as immunomodulation, cell respiration, and protein import[1417]. TSPO is widely distributed throughout the body under physiological conditions. Its expression in the CNS, mainly in glial cells, is upregulated during neuroinflammation[18], which makes it a reliable biomarker to reveal the activation of glial cells[19].

    11C-PK11195: 11C-PK11195 was firstly developed as a TSPO tracer in 1984[20] based on the increased binding of Ro5-4864 and PK11195 in microglial activation. PET using 11C-PK11195 has provided many information on glial activation in many different neurologic disorders, such as AD and PD[2122]. A recent study shows multiple sclerosis patients and healthy controls have the same 11C-PK11195 plasma metabolization rate[23]. Although it can provide many information about microglia activation, its low signal-to-background ratio and high intra-subject variability cause a low sensitivity, thus hampering its clinical utility[2425]. Based on this, to improve specific binding to TSPO and sensitivity, many second-generation tracers were developed.

    11C-DPA-713: 11C-DPA-713 (a pyrazolopyrimidine ligand) is one of the most promising second- generation TSPO traces with low lipophilicity and good affinity for TSPO[26]. It displays a higher signal-to-noise ratio than 11C-PK11195 with lower unspecific binding in vivo[27]. Its larger brain signal is proved in human studies[28]. Recent studies have shown that 11C-DPA-713 kinetic modeling possesses properties suitable for TSPO quantification with PET imaging[2930]. Other pyrazolopyrimidine tracers developed from 11C-DPA-713, such as 11C-DPA-714[3132], VUIIS1008[33] and compound 7[34] are also under studies.

    11C-PBR28: Developed from 11C-DAA1097 and 11C-DAA1106[35], 11C-PBR28 is an aryloxyanilide ligand with high affinity for TSPO[36]. Similar to 11C-DPA-713, 11C-PBR28 has a higher specific binding in human than 11C-PK11195[37]. A test in AD patients showed that 11C-PBR28 level changes with disease progressing and may be useful in longitudinal studies of AD[38]. It can also reveal the extensive glial activation in patients with chronic MCA stroke[39] and semantic dementia[40].

    The second-generation TSPO tracer has a common limitation that hampers their use in clinical application— the sensitivity to a common polymorphism (rs6971), which means that subjects with same TSPO density may produce different PET signals[41]. Therefore, here goes the third generation TSPO PET tracers that may solve this difficulty and keep patients free from a pre-genotyping of TSPO.

    11C-ER176: 11C-ER176 is an isoquinoline analog ligand with a similar structure to 11C-PK11195[42], which shows an adequate sensitivity to robustly image all three affinity genotypes. Although it still gives a lower signal in low-affinity gene type patients compared to high-affinity and missed-affinity patients, it’s high enough to be distinguished from non-specific tissue uptake[43]. 11C-ER176 also produces no radiometabolites that can pass through blood-brain-barrier (BBB)[44]. It has been successfully used to evaluate microglial activation in relation to neurodegenerative progression[45].

    18F-GE180: 18F-GE180 (also known as 18F-flutriciclamide) is another third-generation tracer with low sensitivity to rs6971[4647]. It also shows a better signal-to-noise ratio and lower nonspecific binding. However, a recent study has shown that the advantages may be illusory due to its poor image quality and lack of ability to cross the BBB[48].

    11C-L-deprenyl: 11C-L-deprenyl is the most wildly studied non-TSPO radioligand targeting to monoamine oxidase B (MAO-B)[49]. The PET radioligand version of l-deprenyl or selegiline performs as a selective irreversible MAO-B inhibitor and binds with MAO-B enzyme in the temporal and the white matter in AD patients[50], which indicates its ability to serve as a reliable PET tracer for imaging neuroinflammation .

    In the aspect of glial cell imaging, the specificity of TSPO PET for revealing microglial activation has been fully established. From the first generation TSPO PET tracer to the latest third generation, a higher sensitivity and lower off-target binding has been achieved, together with an adequately high binding for all rs6971 genotypes. Alternative strategies for the synthesis of the third-generation tracers have been studied[45,51]. Many other novel tracers based on them are also being tested[52].

    In neuroinflammation response, both TSPO and MAO-B are just one aspect that may reflect the immune progress. There are many other biomarkers that can be used to study neuroinflammation, such as glycogen synthase kinase 3 (GSK-3), reactive oxygen species (ROS), imidazoline-2 binding sites (I2BS), cyclooxygenase (COX), sphingosine-1-phosphate receptor 1 (S1P1), cannabinoid receptor 2 (CB2R), C-X3-C motif chemokine receptor 1 (CX3CR1), P2X ligand-gated ion channel type 7 (P2X7R), purinergic metabotropic 12 receptor (P2Y12R), receptor for advanced glycation endproducts (RAGE), Mer proto-oncogene tyrosine kinase (MerTK), and triggering receptor expressed on myeloid cells-1 (TREM1)[18,53]. For example, 11C-JNJ-54173717 is a promising tracer targeting to P2X7R which has been used to study amyotrophic lateral sclerosis[54]. Some of those tracers show more promising results for measuring neuroinflammation than TSPO-targeted tracers, although most of those tracers haven't been able to be studied in clinical trials. Some of the new targets have shown the selectivity between microglia and astrocytes. For instance, MAO-B and I2BS are mainly expressed by astrocytes, P2X7R, ROS, COX, S1P1, P2Y12R and TREM1 are mainly expressed by microglia[53]. PET tracers that selective to glial cells are summarized in Table 1[2030,3640,4244,4650,54].

    Table  1.  A summary for positron emission tomography tracers that selective to glial cells
    Mainly expressed cellsBiomarkersTracers
    Microglia and astrocytes TSPO 11C-PK11195[2025], 11C-DPA-713[2630], 11C-PBR28[3640], 11C-ER176[4244], 18F-GE180[4648]
    GSK-3 11C-SB-216763
    CB2R 18F-MA3
    MerTK 18F-HU16907
    Astrocytes MAO-B 11C-L-deprenyl[4950]
    I2BS 11C-BU99008
    Microglia P2X7R JNJ-54173717[54]
    ROS 11C-ascorbicacid
    COX 11C-PS13
    S1P1 18F-TZ4881
    P2Y12R 11C-2
    TREM1 64Cu-TREM1-mAb
    18F: 18fluorine; 11C: 11carbon; 64Cu: 64cuprum; TSPO: translocator protein; GSK-3: glycogen synthase kinase 3; CB2R: cannabinoid receptor 2; MerTK: Mer tyrosine kinase; MAO-B: monoamine oxidase B; I2BS: imidazoline-2 binding sites; P2X7R: P2X ligand-gated ion channel type 7; ROS: reactive oxygen species; COX: cyclooxygenase; S1P1: sphingosine-1-phosphate receptor 1; P2Y12R: purinergic metabotropic 12 receptor; TREM1: triggering receptor expressed on myeloid cells-1.
     | Show Table
    DownLoad: CSV

    WHO classification uses molecular parameters in addition to histology for glioma classification, which indicates that non-invasive molecular imaging technic focusing on depicting metabolic processes, such as PET, may be more widely used. In clinical practice, conventional MRI is mostly used for evaluation of gliomas. However, MRI imaging is short of pathology-specific biomarkers and limited by overall diagnostic and prognostic efficacy[55]. In the contrary, as a functional molecular imaging, PET can provide additional information about metabolism processes, which are useful for primary and differential diagnosis, delineation of glioma extent, grading, treatment planning, assessment of treatment response and prognosis in gliomas. Currently, more than 70 different PET tracers have been used in brain tumor examinations[56]. In this section, we enumerate some representative tracers with the potential of clinical practice.

    As a glucose analogue, 18F-fluorodeoxyglucose (18F-FDG) is the most commonly used radiotracer and metabolizes in cells similar to glucose, which makes it possible to use 18F-FDG to reflect the glucose metabolism. Abnormal proliferation of cancer cells leads to increased glycolysis, resulting in massive accumulation of 18F-FDG. However, the poor tumor-to-background contrast due to the high level of glucose metabolism in normal brain tissue hampers the use of 18F-FDG in brain tumors. The differential diagnosis between glioma and other brain tumor, such as CNS lymphoma, is also limited because of their similar metabolic profiles[57] and overlap of maximum standard uptake values (SUVmax)[58]. It's also difficult for 18F-FDG-PET to distinguish between glioma and some non-neoplastic diseases such as stroke, MS, and infectious diseases, which are also associated with hypermetabolism[59].

    The malignant proliferation of glioma increases the demand for amino acids and accelerates the rate of metabolism[60]. Their increased uptake is caused by the increased transport via the L-type amino acid transporter (LAT) system, especially its subtype LAT1[61]. Different than the high distribution of 18F-FDG in normal brain tissues, the uptake of amino acid is relatively low, so that gliomas can be distinguished from the low background noise. 11C-methyl-L-methionine (11C-MET), O-(2-18F-fluoroethyl)-L-tyrosine (18F-FET) and 3,4-dihydroxy-6-18F-fluoro-L-phenylalanine (18F-DOPA) are widely used radiolabeled amino acid. They are similar and have the ability of passing through intact BBB, the capacity to differentiate tumor recurrence and treatment effects such as pseudo progression and radiation necrosis after treatment.

    11C-MET is one of the most popular amino acid tracers in glioma PET imaging for its convenient production[6263]. The uptake of 11C–MET in cells is mediated by the neutral L-amino acid transporter which serves the increased amino acids utilization of tumor[64]. However, the short half-time of 11C (20.38 minutes) limits its use only in centers with an on-site cyclotron. Glaudemans et al[65] reviewed the value of 11C-MET-PET in imaging gliomas. 11C-MET has high sensitivity (76%–100%) and high specificity (75%–100%) in diagnosis. It can also be used in tumor delineation with improved diagnose accordance rate, compared to CT and MRI. In biopsy and radiotherapy planning, 11C-MET can provide additional information about tumor localization and target volume. A Meta-analysis[66] shows that 11C-MET performs well in differentiation of tumor progression after treatment.

    18F-FET and 11C-MET provide comparable diagnostic information concerning tumor delineation, biopsy determination and recurrence detection[67]. With a long physical half-life (109.77m), 18F-FET can be popularized to more hospitals. Yang et al[68] show that 18F-FET has an average sensitivity of 92% and specificity of 81% in diagnosing glioma. In dynamic PET, time-activity curves of 18F-FET are different between low-grade glioma and high-grade glioma[6970] and provide more additional information for glioma gradingthan 11C-MET-PET and 18F-FDOPA[7172].

    Xiao et al[73] demonstrated a pooled sensitivity of 0.90 and specificity of 0.75 using 18F-FDOPA for diagnosing gliomas, and a pooled sensitivity of 0.88 and specificity of 0.73 in grading gliomas. However, the small sample size results in inconclusive opinion on its clinical effectiveness for glioma imaging. As an amino-acid PET imaging method, numerous evidence supports its superiority of detecting tumor components, which can be used in tumor recurrence and therapy monitoring, compared to enhanced MRI. However, with contradictory conclusions[7476], the feasibility for 18F-FDOPA to distinguish low-grade glioma and high-grade glioma is yet to be studied. 18F-FDOPA is a precursor of dopamine, which may lead to a high uptake in striatum[61].

    3'-deoxy-3'-18F-fluorothymidine (18F-FLT) is a thymidine analog, which can be retained in proliferating tissues by the action of thymidine kinase 1 and presents high-contrast images of tumors[77]. Early study showed that 18F-FLT can detect a larger tumor volume than gadolinium-enhanced MRI imaging in detecting and characterizing brain tumors and provide complementary information together with 11C-MET-PET and MRI. 18F-FLT can also be used for differentiating recurrent glioma and treatment effects[7880] but has a limited role by offering no advantages over 18F-FDG-PET. 18F-FLT can't pass the intact BBB, which limits its usage in tumor delineation and detecting additional metabolic active tumor tissue beyond MRI[8182].

    Choline is an essential ingredient for all cells to synthesize biological membrane molecules while abnormal choline aggregation in tumor tissues is observed due to the high proliferation of tumors. There are two kinds of radiolabeled choline tracers to monitor membrane phospholipids: 11C-choline (11C-CHO) and 18F-fluorocholine (18F-FHO). Studies with 18F-FHO have focused on the evaluation of prostate cancer[83]. The low uptake of 18F-FHO in normal brain tissues presents a better tumor/normal uptake ratio than FDG-PET[84]. Many studies showed that 11C- and 18F- labeled choline has encouraged results in distinguishing different kinds of brain tumors from metastases and benign lesions[83], evaluating the potential malignancy of glioma[85], guiding stereotactic biopsy sampling[86] and differentiating glioma recurrence[8788].

    Tumor hypoxia is an interesting mechanism in tumor progress, which may cause the resistant to radiotherapy[89]. 18F-fluoromisonidazole (18F-FMISO) is a PET tracer that selectively binds to hypoxic tissues and can be used to measure the hypoxia in glioma and provide information for prognosis[90]. As it is retained by irreversible binding to the thiol-rich metabolic proteins at rates that are inversely proportional to oxygen concentration, 18F-FMISO provides quantitative measurements of hypoxia level[91].

    18F-fluciclovine ( also called anti-18F-FACBC) is a synthetic amino acid that has been approved by Food and Drug Administration (FDA) and European Medicines Agency (EMA) for recurrent prostate cancer. Different with other amino acid PET tracers, such as 11C-MET, 18F-FET and 18F-FDOPA, 18F-fluciclovine can be transported by both LAT1 and alanine-serine-cysteine (ASC), specifically ASCT2[92]. These transporters work as an exchange system that brings in a preferential amino acid at the expense of a less essential one, but do not increase the overall cellular levels of amino acids[93]. Many studies have showed that 18F-fluciclovine can delineate glioma in areas not visualized using MRI and be used in glioma grading[92,9496].

    In highly metabolizing and rapidly growing brain tumors, the lack of abundant glucose and oxygen may lead to metabolism reprogramming to obtain sufficient energy, for example, the use of fatty acid[97]. Induced acetate/acetyl CoA metabolism is highly related to fatty acid synthesis supporting tumor growth[98]. Therefore, many radiolabeled acetates may be used in glioma imaging, such as 1-11C-acetate[99100] (11C-ACE), 2-14C-acetate[101] (14C-ACE), 2-18F-fluoroacetate[102] (18F-FAC) and 18F-fluoropivalic acid[103] (18F-FPIA), a newly designed novel radiotracer based on trimethylacetate.

    Angiogenesis is the process of forming new blood vessels and closely related to tumor growth and progression. The αvβ3 integrin is highly expressed only in activated endothelial cells of tumor neovasculature and can be traced by radiolabeled arginine-glycine-aspartic acid (RGD) peptides and analogs, such as monomeric cyclic 18F-galacto-RGD and 18F-AH111585. With increased receptor-binding affinity and tumor uptake, 2-fluoropropionyl-labeled PEGylated dimeric RGD peptide (18F-FPP(RGD)2) is the one used in glioma[104]. 18F-FPP(RGD)2 has increased uptake in glioblastoma multiforme and would be suitable for clinical examinations[105106].

    18F-FDG is the most widely used PET tracer in oncology but the high uptake in normal brain tissue hampers its clinical use. According to the Response Assessment in Neuro-Oncology group, amino-acid PET is recommended for PET imaging of glioma[107]. It does not seem to be one particular tracer that could be recommended over other tracers in all aspects of imaging glioma, maybe even not in any specific usage 18. More studies with larger cohort and unified standard are necessary to find the best tracer among all the tracers that have already been used in clinical trials for many years. Some newly designed tracers such as 68Ga-Alb-FAPtp-01[108] and 89Zr/177Lu-labeled Lumi804-aCD11b can be used both in imaging and treatment[109]. Tracers for glioma imaging are summarized in Table 2[6263,6590,92,9496,99106].

    Table  2.  A summary for positron emission tomography tracers used for glioma
    Chemical typeTracer  Target or intake mechanismNote
    Glucose analogue 18F-FDG Glucose Demonstrate glucose metabolism;
    Poor tumor-to-background contrast
    Amino-acid 11C-MET[6263,6566] LAT1 Demonstrate amino-acid uptake;
    High sensitivity and specificity;
    Best and most studied tracers for glioma
    18F-FET[6772]
    18F-FDOPA[7376]
    Thymidine analog 18F- FLT[7782] Thymidine kinase 1 Markers of cell proliferation;
    Cannot pass intact BBB
    Choline 11C-CHO[8486,88] Choline transporter Monitor membrane phospholipids;
    Cannot pass intact BBB
    18F-FHO[83,8687]
    Nitroimidazole 18F-FMISO[8990] Nitroreductase enzymes Investigate intratumoural hypoxia;
    Haven't achieved clinical relevance
    Amino acid 18F-fluciclovine [92,9496] LAT1 & ASCT2 Demonstrate amino-acid uptake;
    Transported by both LAT1 and ASCT2
    Acetate 11C-ACE[99100] Acetate/acetyl CoA metabolism Demonstrate the use of fatty acid
    14C-ACE[101]
    18F-FAC[102]
    18F-FPIA[103]
    Peptide 18F-FPP(RGD)2[104106] αvβ3-integrin Demonstrate angiogenesis
    18F: 18fluorine; 11C: 11carbon; FDG: fluorodeoxyglucose; LAT1: L-type amino acid transporter-1; MET: methyl-L-methionine; FET: O-(2-18F-fluoroethyl)-L-tyrosine; FDOPA: 3,4-dihydroxy-6-18F-fluoro-L-phenylalanine; FLT: 3′-deoxy-3′-18F-fluorothymidine; CHO: choline; FHO: fluorocholine; FMISO: fluoromisonidazole; ASCT2: alanine-serine-cysteine-2; ACE: acetate; FAC: fluoroacetate; FPIA: fluoropivalic acid; FPP(RGD)2: 2-fluoropropionyl-labeled PEGylated dimeric RGD peptide; BBB: blood-brain-barrier.
     | Show Table
    DownLoad: CSV

    AD is the most common type of dementia. It makes up about two-third of all differential diagnoses about dementia. About 10% of people elder than 65 are thought to have AD[110111]. As a progressive and irreversible neurodegenerative disorder, patients with AD suffer from gradual loss of memory and progressive cognitive dysfunction involving the language, visuospatial and executive domains. In 1970s, cholinergic hypothesis of AD was proposed according to the essential role of acetylcholine in cognitive function. However, the most accepted hypothesis of AD is amyloid cascade hypothesis[112], which posits that amyloid-β (Aβ) accumulation is the primary event leading to a cascade of effects that result in neuronal damage while hyperphosphorylated protein (e.g., p-Tau) tangles, the second hallmark of AD, is a downstream effect of Aβ accumulation. Therefore developing associated agents for detecting these AD abnormal hallmarks in the brain may be helpful for the early and differential diagnosis of the disease progression and prognosis evaluation.

    The golden-standard diagnosis of AD is based on autopsy while 40% patients may be misdiagnosed under current in vivo clinical diagnosis criteria because of the difficulties in accurate discrimination of AD and non-AD dementia[113]. According to the National Institute on Aging and the Alzheimer's Association, AD could be defined and studied in terms of three stages: preclinical, mild cognitive impairment (MCI) and dementia[114]. It is suggested to prevent AD in the preclinical phase, rather than trying to cure it when clinical symptoms have appeared.

    In AD patient, the balance between production and clearance of Aβ is broken, which causes the abnormal accumulation and result in aggregates with β-pleated sheet structure[115]. Extracellular Aβ oligomers bind the cell surface, leading to receptors' functional of disruption and finally cause the synaptic dysfunction and neurodegeneration[116]. The Aβ accumulation comes far before the clinical symptoms and may be used for early prediction of AD. The level of Aβ in cerebrospinal fluid (CSF) has been recommended as a usable and cost-less tool in the diagnosis and monitoring of AD by the European Medicines Agency[117118]. PET imaging with Aβ targeted radiotracers can hopefully identify potential AD patients and monitor its progression in a non-invasive and safer way.

    11C-PIB: The earliest and most widely used and studied radiotracer for amyloid PET imaging is N-methyl-11C-2-(4′-methylaminophenyl)-6-hydroxybenzothiazole, a benzothiazole derivative, also called 11C-Pittsburgh compound B (11C-PIB)[119]. It is derived from Thioflavin T (Th-T), a histopathological dye that binds to the amyloid plaque, as well as Congo Red. 11C-PIB can easily cross the BBB and bind to amyloid plaques with high affinity. It is proved that 11C-PIB retention in the frontal cortex and posterior cingulum significantly correlate with CSF Aβ[120], and can be used to predict the disease progression from MCI to AD[121123].

    18F-flutemetamol: 18F-flutemetamol (Vizamyl, also known as 18F-GE067 and 3'-18F-PIB) is developed in the seeking of 18F labeled radiotracer similar to 11C-PIB but can be popularize to centers without cyclotron. Many studies have shown that the uptake processes and specific binding characteristics of flutemetamol is very similar to 11C-PIB[124125]. It has been approved by FDA in 2013.

    18F-florbetapir and 18F-florbetaben: 18F-florbetapir (Amyvid, also known as 18F-AV45 or florpiramine) and 18F-florbetaben (Neuraceq, also known as 18F-AV-1 or 18F-BAY94-9172) are both diaryl alkene analogs. In this branch, 3H-SB-13 is the first stilbene derivative with high affinity to Aβ[126], and then led to its 11C-labeled analog 11C-SB-13[127]. 18F-florbetapir is the first 18F-labeled PET tracer developed for imaging amyloid plaques and has been approved by FDA in 2012. 18F-florbetaben was approved by FDA in 2014.

    The three FDA approved amyloid tracers[113] of 18F-flutemetamol, 18F-florbetapir and 18F-florbetaben all show high correlation with 11C-PIB and can be used in the prediction of MCI progression and diagnosis of AD. Since nearly all the studies about those tracers are compared with 11C-PIB, it is hard to deny the hypothesis that they are quite similar in sensitivity and specificity. The biggest problem of the first generation of Aβ pet tracers is the low signal-to-background ratio, which limits the sensitivity especially in the early phase AD patients with low plaque levels. Hence, many second-generation tracers were developed to solve this problem.

    18F-flutafuranol: 18F-flutafuranol (also known as 18F-AZD4694 or 18F-NAV4694) is a benzofuran derivative firstly synthesized in 2010[126]. Same as the development of 18F-florbetapir and 18F-florbetaben, the first tracer in this branch was also 3H labeled, 3H-AZD2184, which displays a twice higher signal-to-background ratio than 3H-PIB[128]. The high signal-to-background ratio is also observed on 11C-AZD2184[129]. 18F-flutafuranol shares the identical binding with the second-generation tracers and shows a lower non-specific binding in vitro. Following studies show that 18F-flutafuranol has a high cortical binding in AD, highly correlating to 11C-PIB, and lower non-specific white matter binding, which confirmed its potential for wide clinical application[130131].

    According to the amyloid cascade hypothesis, the generation of Aβ plaques is the primary progression of AD and causes other downstream events, such as Tau aggregates. However, amyloid volume has no additional value on disease severity[132]. On the contrary, the extent of neurofibrillary tangles composed of Tau, seems to be increased with disease progression and is closely related to memory decline[133]. Therefore, Tau PET is used for both early diagnosis and tracing disease progression.

    18F-FDDNP: 18F-FDDNP is the first PET tracer labelling regional Tau tangles and shows the ability of differentiating MCI from normal aging and AD[134]. However, its relatively high affinity to Aβ limits the Tau imaging specifically[135]. In order to improve the specificity and selectivity, many other sets of tracers were synthetized and referred to as the first generation.

    Quinoline derivatives [2-(4-aminophenyl)-6-(2-fluoroethoxy)quinoline] [THK] series): Based on the new quinoline and benzimidazole derivatives that bind Tau over Aβ plaques found by Okamura et al[136], the first tracer of the THK series of 18F-THK523 was developed and fulfils ligand criteria for human imaging trials[137]. Afterwards, 18F-THK5105 and 18F-THK5117 are found to have a higher binding affinity than 18F-THK523[138] but have a relative high retention within the white matter[139]. The latest tracer in this series is 18F-THK-5351 (also known as GE-216) with faster kinetics, higher contrast, and lower retention in subcortical white matter than18F-THK5117[140]. Many studies have shown that the THK series have a high off-target binging to MAO-B, which accumulates with age, and hence, may be limited in the use of Tau specific imaging.

    The carbazole and benzimidazole derivatives (18F-flortaucipir): 18F-flortaucipir (also known as Tauvid, 18F-AV1451 or 18F-T807) is a benzimidazole pyrimidine derivatives developed in 2013 and has high affinity, selectivity, and lipophilicity[141]. It provides high contrast because of low retention in white matter. Clinical studies show that 18F-flortaucipir PET present significant correlations with CSF measurements of tau pathology[142] and is associated with clinical impairment[143]. 18F-flortaucipir has been approved by FDA for Tau imaging in 2020. However, 18F-flortaucipir also has similar affinity to MAO-B, like the THK-series[144].

    11C-PBB3: 11C-PBB3 (pyridinyl-butandienyl-benzothiazole 3) is another tracer for Tau[145]. 11C-PBB3 PET has a robust signal in Tau enriched tissue and is consistent with the spreading of Tau pathology with AD progression. It also has a low white matter binding similar as 18F-flortaucipi while the radiometabolites may remain in the brain and hamper its quantification.

    There are some common limits in the first generation of Tau tracers, such as retention in white matter and off-target binding especially to MAO-B. Therefore, the second generation now enters the field. Some of them are improved from reported tracers and have a similar structure as the first generation.

    Subsequent 18F-flortaucipir series: Based on 18F-T808 (also called 18F-AV680), which is in the same series with 18F-flortaucipir but abandoned for its propensity to metabolic defluorination[146], 18F-GTP1 (18F-Genentech tau probe 1) was developed without measurable binding to MAO-B and evidence of defluorination[147]. Based on 18F-flortaucipir, three compounds were developed with lower affinity to MAO-B than 18F-flortaucipir and THK-series, in which 18F-RO948 (18F-RO6958948) was found to have a better pharmacokinetics and metabolic properties and higher signal-to-background ratio[148]. 18F-PI-2620 is another Tau tracer improved from 18F-flortaucipir without off-target binding to MAO-B[149].

    18F-PM-PBB3: Based on 11C-PBB3, 18F-PM-PBB3 (18F-APN-1607) has developed with higher metabolic stability and less off-target signals, which makes it higher signal-to-background ratio[150]. 18F-PM-PBB3 also correlated well with cognitive changes[151].

    18F-MK6240: 18F-MK6240 is an azaindole derivatives developed in 2016[152]. It can efficiently differentiate normal control group, MCI and AD patients. No defluorination or off-target signals to MAO-B has been observed[152]. Clinical trials also showed a minimal off-target binding in human brain[153].

    18F-JNJ311: 18F-JNJ311 (18F-JNJ64349311) is a naphthyridine derivatives developed in 2017[154]. Animal studies showed its moderate initial brain uptake, rapid brain washout, and minor off-target specific binding in the healthy rat brain[155]. 18F-JNJ311 has a favorable pharmacokinetic profile like 18F-flortaucipir and is a potential candidate for Tau-specific tracer.

    Tracers for Alzheimer's disease are summarized in Table 3[113,119126,128131,134135,137142,144145,147155].

    Table  3.  A summary for positron emission tomography tracers used for Alzheimer's disease
    TargetGenerationTracersNote
    Planar heteroaromatic analogs 1 11C-PIB[119123] Classical used tracer
    1 18F-flutemetamol[124125] FDA approved
    1 18F-florbetapir[113] FDA approved
    1 18F-florbetaben[113] FDA approved
    2 18F-flutafuranol[126,128131]
    Tau
    Naphthalene analogs 0 18F-FDDNP[134135] Affinity for both Aβ and Tau
    Arylquinoline derivatives (THK series) 1 18F-THK-523[137]
    1 18F-THK-5105[138]
    1 18F-THK-5117[138]
    1 18F-THK-5351[140]
    Benzimidazole pyrimidines derivatives 1 18F-flortaucipir[141142,144] FDA approved
    2 18F-RO948[148]
    2 18F-PI2620[149]
    2 18F-GTP1[147]
    PBB 1 11C-PBB3[145]
    2 18F-PM-PBB3[150151]
    Azaindole derivatives 2 18F-MK6240[152153]
    Naphthyridine derivatives 2 18F-JNJ311[154155]
    Aβ: amyloid-β protein; 18F: 18fluorine; 11C: 11carbon; PIB: Pittsburgh compound B; FDDNP: 2-(1-{6-[(2-fluoroethyl)(methyl)amino]-2-naphthyl}ethylidene)malononitrile; PBB3: pyridinyl-butadienyl-benzothiazole 3; FDA: Food and Drug Administration.
     | Show Table
    DownLoad: CSV

    There are many hurdles in developing PET tracers for glial cells and related disease. For example, a candidate tracer must have the ability to across BBB. After entering the CNS, it must have sufficient affinity and specificity to its target with low off-target binding. In the field of glial tracer imaging, future work should focus on the different phenotypes of microglia and astrocytes. It's clear that there is more complex and precision differentiation, which can be a great progress to understand the precise roles of glial cells in neuroinflammation and many other related diseases. Some advanced technology can be helpful for development of new glial tracers, such as computational BBB permeability modeling tools, human BBB microfluidic and BBB organoids.

    Furthermore, new imaging systems may bring a revelation of glial imaging with better resolution and sensitivity. PET/MRI is a technology that combines the anatomic and quantitative strengths of MRI with physiologic information obtained from PET. Compared with PET/CT, it has an increased soft tissue contrast and lacks of ionizing radiation exposure, which outperforms PET/CT in many applications[156]. It has been used in both neurodegenerative disorders[157159] and glioma[160162], with above-mentioned radiotracers, PET/MRI has achieved much better results than using PET or MRI individually, and also PET/CT.

    This work was supported by the Key Laboratory of Biomedical Engineering of Hainan Province under Opening Foundation 2022001.

    CLC number: R741.04; R817.4, Document code: A

    The authors reported no conflict of interests.

  • [1]
    Von Bartheld CS. Myths and truths about the cellular composition of the human brain: a review of influential concepts[J]. J Chem Neuroanat, 2018, 93: 2–15. doi: 10.1016/j.jchemneu.2017.08.004
    [2]
    Herculano-Houzel S. The glia/neuron ratio: how it varies uniformly across brain structures and species and what that means for brain physiology and evolution[J]. Glia, 2014, 62(9): 1377–1391. doi: 10.1002/glia.22683
    [3]
    Jäkel S, Dimou L. Glial cells and their function in the adult brain: a journey through the history of their ablation[J]. Front Cell Neurosci, 2017, 11: 24. doi: 10.3389/fncel.2017.00024
    [4]
    Nayak D, Roth TL, McGavern DB. Microglia development and function[J]. Annu Rev Immunol, 2014, 32: 367–402. doi: 10.1146/annurev-immunol-032713-120240
    [5]
    Siracusa R, Fusco R, Cuzzocrea S. Astrocytes: role and functions in brain pathologies[J]. Front Pharmacol, 2019, 10: 1114. doi: 10.3389/fphar.2019.01114
    [6]
    Disabato DJ, Quan N, Godbout JP. Neuroinflammation: the devil is in the details[J]. J Neurochem, 2016, 139(S2): 136–153. doi: 10.1111/jnc.13607
    [7]
    Mantovani A, Sica A, Locati M. Macrophage polarization comes of age[J]. Immunity, 2005, 23(4): 344–346. doi: 10.1016/j.immuni.2005.10.001
    [8]
    Zamanian JL, Xu L, Foo LC, et al. Genomic analysis of reactive astrogliosis[J]. J Neurosci, 2012, 32(18): 6391–6410. doi: 10.1523/JNEUROSCI.6221-11.2012
    [9]
    Lyman M, Lloyd DG, Ji X, et al. Neuroinflammation: the role and consequences[J]. Neurosci Res, 2014, 79: 1–12. doi: 10.1016/j.neures.2013.10.004
    [10]
    Pandey R, Caflisch L, Lodi A, et al. Metabolomic signature of brain cancer[J]. Mol Carcinog, 2017, 56(11): 2355–2371. doi: 10.1002/mc.22694
    [11]
    Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization classification of tumors of the central nervous system: a summary[J]. Acta Neuropathol, 2016, 131(6): 803–820. doi: 10.1007/s00401-016-1545-1
    [12]
    Walker C, Baborie A, Crooks D, et al. Biology, genetics and imaging of glial cell tumours[J]. Br J Radiol, 2011, 84(S2): S90–S106. doi: 10.1259/bjr/23430927
    [13]
    Santoro A, Mattace Raso G, Taliani S, et al. TSPO-ligands prevent oxidative damage and inflammatory response in C6 glioma cells by neurosteroid synthesis[J]. Eur J Pharm Sci, 2016, 88: 124–131. doi: 10.1016/j.ejps.2016.04.006
    [14]
    Veenman L, Gavish M. The peripheral-type benzodiazepine receptor and the cardiovascular system. Implications for drug development[J]. Pharmacol Ther, 2006, 110(3): 503–524. doi: 10.1016/j.pharmthera.2005.09.007
    [15]
    Hauet T, Yao Z, Bose HS, et al. Peripheral-type benzodiazepine receptor-mediated action of steroidogenic acute regulatory protein on cholesterol entry into leydig cell mitochondria[J]. Mol Endocrinol, 2005, 19(2): 540–554. doi: 10.1210/me.2004-0307
    [16]
    O’Hara MF, Nibbio BJ, Craig RC, et al. Mitochondrial benzodiazepine receptors regulate oxygen homeostasis in the early mouse embryo[J]. Reprod Toxicol, 2003, 17(4): 365–375. doi: 10.1016/S0890-6238(03)00035-2
    [17]
    Larcher JC, Vayssiere JL, Le Marquer FJ, et al. Effects of peripheral benzodiazepines upon the O2 consumption of neuroblastoma cells[J]. Eur J Pharmacol, 1989, 161(2–3): 197–202. doi: 10.1016/0014-2999(89)90843-1
    [18]
    Jain P, Chaney AM, Carlson ML, et al. Neuroinflammation PET imaging: current opinion and future directions[J]. J Nucl Med, 2020, 61(8): 1107–1112. doi: 10.2967/jnumed.119.229443
    [19]
    Nutma E, Ceyzériat K, Amor S, et al. Cellular sources of TSPO expression in healthy and diseased brain[J]. Eur J Nucl Med Mol Imaging, 2021, 49(1): 146–163. doi: 10.1007/s00259-020-05166-2
    [20]
    Camsonne R, Crouzel C, Comar D, et al. Synthesis of N-(11C) methyl, N-(methyl-1 propyl), (chloro-2 phenyl)-1 isoquinoleine carboxamide-3 (PK 11195): a new ligand for peripheral benzodiazepine receptors[J]. J Label Compd Radiopharm, 1984, 21(10): 985–991. doi: 10.1002/jlcr.2580211012
    [21]
    Cagnin A, Brooks DJ, Kennedy AM, et al. In-vivo measurement of activated microglia in dementia[J]. Lancet, 2001, 358(9280): 461–467. doi: 10.1016/S0140-6736(01)05625-2
    [22]
    Ouchi Y, Yoshikawa E, Sekine Y, et al. Microglial activation and dopamine terminal loss in early Parkinson's disease[J]. Ann Neurol, 2005, 57(2): 168–175. doi: 10.1002/ana.20338
    [23]
    De Souza AM, Pitombeira MS, De Souza LE, et al. 11C-PK11195 plasma metabolization has the same rate in multiple sclerosis patients and healthy controls: a cross-sectional study[J]. Neural Regen Res, 2021, 16(12): 2494–2498. doi: 10.4103/1673-5374.313062
    [24]
    Jučaite A, Cselényi Z, Arvidsson A, et al. Kinetic analysis and test-retest variability of the radioligand [11C](R)-PK11195 binding to TSPO in the human brain-a PET study in control subjects[J]. EJNMMI Res, 2012, 2: 15. doi: 10.1186/2191-219X-2-15
    [25]
    Parente A, Feltes PK, Vallez García D, et al. Pharmacokinetic analysis of 11C-PBR28 in the rat model of herpes encephalitis: comparison with (R)-11C-PK11195[J]. J Nucl Med, 2016, 57(5): 785–791. doi: 10.2967/jnumed.115.165019
    [26]
    James ML, Fulton RR, Henderson DJ, et al. Synthesis and in vivo evaluation of a novel peripheral benzodiazepine receptor PET radioligand[J]. Bioorg Med Chem, 2005, 13(22): 6188–6194. doi: 10.1016/j.bmc.2005.06.030
    [27]
    Boutin H, Chauveau F, Thominiaux C, et al. 11C-DPA-713: a novel peripheral benzodiazepine receptor PET ligand for in vivo imaging of neuroinflammation[J]. J Nucl Med, 2007, 48(4): 573–581. doi: 10.2967/jnumed.106.036764
    [28]
    Endres CJ, Pomper MG, James M, et al. Initial evaluation of 11C-DPA-713, a novel TSPO PET ligand, in humans[J]. J Nucl Med, 2009, 50(8): 1276–1282. doi: 10.2967/jnumed.109.062265
    [29]
    Yasuno F, Kimura Y, Ogata A, et al. Kinetic modeling and non-invasive approach for translocator protein quantification with 11C-DPA-713[J]. Nucl Med Biol, 200, 108–109: 76–84. doi: 10.1016/j.nucmedbio.2022.02.005
    [30]
    Akerele MI, Zein SA, Pandya S, et al. Population-based input function for TSPO quantification and kinetic modeling with [11C]-DPA-713[J]. EJNMMI Phys, 2021, 8(1): 39. doi: 10.1186/s40658-021-00381-8
    [31]
    Sarda-Mantel L, Alsac JM, Boisgard R, et al. Comparison of 18F-fluoro-deoxy-glucose, 18F-fluoro-methyl-choline, and 18F-DPA714 for positron-emission tomography imaging of leukocyte accumulation in the aortic wall of experimental abdominal aneurysms[J]. J Vasc Surg, 2012, 56(3): 765–773. doi: 10.1016/j.jvs.2012.01.069
    [32]
    Kaneko KI, Irie S, Mawatari A, et al. [18F]DPA-714 PET imaging for the quantitative evaluation of early spatiotemporal changes of neuroinflammation in rat brain following status epilepticus[J]. Eur J Nucl Med Mol Imaging, 2022, 49(7): 2265–2275. doi: 10.1007/s00259-022-05719-7
    [33]
    Tang D, McKinley ET, Hight MR, et al. Synthesis and structure-activity relationships of 5, 6, 7-substituted pyrazolopyrimidines: discovery of a novel TSPO PET ligand for cancer imaging[J]. J Med Chem, 2013, 56(8): 3429–3433. doi: 10.1021/jm4001874
    [34]
    Li J, Smith JA, Dawson ES, et al. Optimized translocator protein ligand for optical molecular imaging and screening[J]. Bioconjugate Chem, 2017, 28(4): 1016–1023. doi: 10.1021/acs.bioconjchem.6b00711
    [35]
    Okubo T, Yoshikawa R, Chaki S, et al. Design, synthesis and structure–affinity relationships of aryloxyanilide derivatives as novel peripheral benzodiazepine receptor ligands[J]. Bioorg Med Chem, 2004, 12(2): 423–438. doi: 10.1016/j.bmc.2003.10.050
    [36]
    Briard E, Zoghbi SS, Imaizumi M, et al. Synthesis and evaluation in monkey of two sensitive 11C-labeled aryloxyanilide ligands for imaging brain peripheral benzodiazepine receptors in vivo[J]. J Med Chem, 2008, 51(1): 17–30. doi: 10.1021/jm0707370
    [37]
    Kreisl WC, Fujita M, Fujimura Y, et al. Comparison of [11C]-(R)-PK 11195 and [11C]PBR28, two radioligands for translocator protein (18 kDa) in human and monkey: Implications for positron emission tomographic imaging of this inflammation biomarker[J]. Neuroimage, 2010, 49(4): 2924–2932. doi: 10.1016/j.neuroimage.2009.11.056
    [38]
    Nair A, Veronese M, Xu X, et al. Test-retest analysis of a non-invasive method of quantifying [11C]-PBR28 binding in Alzheimer's disease[J]. EJNMMI Res, 2016, 6(1): 72. doi: 10.1186/s13550-016-0226-3
    [39]
    Schaechter JD, Hightower BG, Kim M, et al. A pilot [11C]PBR28 PET/MRI study of neuroinflammation and neurodegeneration in chronic stroke patients[J]. Brain Behav Immun Health, 2021, 17: 100336. doi: 10.1016/j.bbih.2021.100336
    [40]
    Pascual B, Funk Q, Zanotti-Fregonara P, et al. Neuroinflammation is highest in areas of disease progression in semantic dementia[J]. Brain, 2021, 144(5): 1565–1575. doi: 10.1093/brain/awab057
    [41]
    Owen DR, Yeo AJ, Gunn RN, et al. An 18-kDa translocator protein (TSPO) polymorphism explains differences in binding affinity of the PET radioligand PBR28[J]. J Cereb Blood Flow Metab, 2012, 32(1): 1–5. doi: 10.1038/jcbfm.2011.147
    [42]
    Zanotti-Fregonara P, Zhang Y, Jenko KJ, et al. Synthesis and evaluation of translocator 18 kDa protein (TSPO) positron emission tomography (PET) radioligands with low binding sensitivity to human single nucleotide polymorphism rs6971[J]. ACS Chem Neurosci, 2014, 5(10): 963–971. doi: 10.1021/cn500138n
    [43]
    Ikawa M, Lohith TG, Shrestha S, et al. 11C-ER176, a Radioligand for 18-kDa translocator protein, has adequate sensitivity to robustly image all three affinity genotypes in human brain[J]. J Nucl Med, 2017, 58(2): 320–325. doi: 10.2967/jnumed.116.178996
    [44]
    Fujita M, Kobayashi M, Ikawa M, et al. Comparison of four 11C-labeled PET ligands to quantify translocator protein 18 kDa (TSPO) in human brain: (R)-PK11195, PBR28, DPA-713, and ER176-based on recent publications that measured specific-to-non-displaceable ratios[J]. EJNMMI Res, 2017, 7(1): 84. doi: 10.1186/s13550-017-0334-8
    [45]
    Rocha NP, Charron O, Latham LB, et al. Microglia activation in basal ganglia is a late event in huntington disease pathophysiology[J]. Neurol Neuroimmunol Neuroinflamm, 2021, 8(3): e984. doi: 10.1212/NXI.0000000000000984
    [46]
    Boutin H, Murray K, Pradillo J, et al. 18F-GE-180: a novel TSPO radiotracer compared to 11C-R-PK11195 in a preclinical model of stroke[J]. Eur J Nucl Med Mol Imaging, 2015, 42(3): 503–511. doi: 10.1007/s00259-014-2939-8
    [47]
    Vettermann FJ, Harris S, Schmitt J, et al. Impact of TSPO receptor polymorphism on [18F]GE-180 binding in healthy brain and pseudo-reference regions of neurooncological and neurodegenerative disorders[J]. Life (Basel), 2021, 11(6): 484. doi: 10.3390/life11060484
    [48]
    Zanotti-Fregonara P, Pascual B, Rostomily RC, et al. Anatomy of 18F-GE180, a failed radioligand for the TSPO protein[J]. Eur J Nucl Med Mol Imaging, 2020, 47(10): 2233–2236. doi: 10.1007/s00259-020-04732-y
    [49]
    Lammertsma AA, Bench CJ, Price GW, et al. Measurement of cerebral monoamine oxidase B activity using L-[11C]deprenyl and dynamic positron emission tomography[J]. J Cereb Blood Flow Metab, 1991, 11(4): 545–556. doi: 10.1038/jcbfm.1991.103
    [50]
    Gulyás B, Pavlova E, Kása P, et al. Activated MAO-B in the brain of Alzheimer patients, demonstrated by [11C]-L-deprenyl using whole hemisphere autoradiography[J]. Neurochem Int, 2011, 58(1): 60–68. doi: 10.1016/j.neuint.2010.10.013
    [51]
    Mixdorf JC, Murali D, Xin Y, et al. Alternative strategies for the synthesis of [11C]ER176 for PET imaging of neuroinflammation[J]. Appl Radiat Isot, 2021, 178: 109954. doi: 10.1016/j.apradiso.2021.109954
    [52]
    Lee JH, Simeon FG, Liow JS, et al. In vivo evaluation of six analogs of 11C-ER176 as candidate 18F-labeled radioligands for translocator protein 18 kDa (TSPO)[J]. J Nucl Med, 2022, 63(8): 1252–1258. doi: 10.2967/jnumed.121.263168
    [53]
    Narayanaswami V, Dahl K, Bernard-Gauthier V, et al. Emerging PET radiotracers and targets for imaging of neuroinflammation in neurodegenerative diseases: outlook beyond TSPO[J]. Mol Imaging, 2018, 17: 1536012118792317. doi: 10.1177/1536012118792317
    [54]
    Van Weehaeghe D, Van Schoor E, De Vocht J, et al. TSPO versus P2X7 as a target for neuroinflammation: an in vitro and in vivo study[J]. J Nucl Med, 2020, 61(4): 604–607. doi: 10.2967/jnumed.119.231985
    [55]
    Upadhyay N, Waldman AD. Conventional MRI evaluation of gliomas[J]. Br J Radiol, 2011, 84(S2): S107–S111. doi: 10.1259/bjr/65711810
    [56]
    Langen KJ, Galldiks N, Hattingen E, et al. Advances in neuro-oncology imaging[J]. Nat Rev Neurol, 2017, 13(5): 279–289. doi: 10.1038/nrneurol.2017.44
    [57]
    Rosenfeld SS, Hoffman JM, Coleman RE, et al. Studies of primary central nervous system lymphoma with fluorine-18-fluorodeoxyglucose positron emission tomography[J]. J Nucl Med, 1992, 33(4): 532–536. https://pubmed.ncbi.nlm.nih.gov/1552337/
    [58]
    Kosaka N, Tsuchida T, Uematsu H, et al. 18F-FDG PET of common enhancing malignant brain tumors[J]. AJR Am J Roentgenol, 2008, 190(6): W365–W369. doi: 10.2214/AJR.07.2660
    [59]
    Omuro AMP, Leite CC, Mokhtari K, et al. Pitfalls in the diagnosis of brain tumours[J]. Lancet Neurol, 2006, 5(11): 937–948. doi: 10.1016/S1474-4422(06)70597-X
    [60]
    Chung JK, Kim YK, Kim SK, et al. Usefulness of 11C-methionine PET in the evaluation of brain lesions that are hypo- or isometabolic on 18F-FDG PET[J]. Eur J Nucl Med Mol Imaging, 2002, 29(2): 176–182. doi: 10.1007/s00259-001-0690-4
    [61]
    Galldiks N, Langen KJ. Applications of PET imaging of neurological tumors with radiolabeled amino acids[J]. Quart J Nucl Med Mol Imaging, 2015, 59(1): 70–82. https://pubmed.ncbi.nlm.nih.gov/25517079/
    [62]
    Långström B, Antoni G, Gullberg P, et al. Synthesis of L- and D-[methyl-11C]methionine[J]. J Nucl Med, 1987, 28(6): 1037–1040. https://pubmed.ncbi.nlm.nih.gov/3585494/
    [63]
    Singhal T, Narayanan TK, Jain V, et al. 11C-L-methionine positron emission tomography in the clinical management of cerebral gliomas[J]. Mol Imaging Biol, 2008, 10(1): 1–18. doi: 10.1007/s11307-007-0115-2
    [64]
    Deuschl C, Kirchner J, Poeppel TD, et al. 11C–MET PET/MRI for detection of recurrent glioma[J]. Eur J Nucl Med Mol Imaging, 2018, 45(4): 593–601. doi: 10.1007/s00259-017-3916-9
    [65]
    Glaudemans AWJW, Enting RH, Heesters MAAM, et al. Value of 11C-methionine PET in imaging brain tumours and metastases[J]. Eur J Nucl Med Mol Imaging, 2013, 40(4): 615–635. doi: 10.1007/s00259-012-2295-5
    [66]
    De Zwart PL, Van Dijken BR J, Holtman GA, et al. Diagnostic accuracy of PET tracers for the differentiation of tumor progression from treatment-related changes in high-grade glioma: a systematic review and metaanalysis[J]. J Nucl Med, 2020, 61(4): 498–504. doi: 10.2967/jnumed.119.233809
    [67]
    Grosu AL, Astner ST, Riedel E, et al. An interindividual comparison of O-(2-[18F]fluoroethyl)-L-tyrosine (FET)- and L-[methyl-11C]methionine (MET)-PET in patients with brain gliomas and metastases[J]. Int J Radiat Oncol Biol Phys, 2011, 81(4): 1049–1058. doi: 10.1016/j.ijrobp.2010.07.002
    [68]
    Yang Y, He MZ, Li T, et al. MRI combined with PET-CT of different tracers to improve the accuracy of glioma diagnosis: a systematic review and meta-analysis[J]. Neurosurg Rev, 2019, 42(2): 185–195. doi: 10.1007/s10143-017-0906-0
    [69]
    Weckesser M, Langen KJ, Rickert CH, et al. O-(2-[18F]fluorethyl)-L-tyrosine PET in the clinical evaluation of primary brain tumours[J]. Eur J Nucl Med Mol Imaging, 2005, 32(4): 422–429. doi: 10.1007/s00259-004-1705-8
    [70]
    Pöpperl G, Kreth FW, Herms J, et al. Analysis of 18F-FET PET for grading of recurrent gliomas: is evaluation of uptake kinetics superior to standard methods?[J]. J Nucl Med, 2006, 47(3): 393–403. https://pubmed.ncbi.nlm.nih.gov/16513607/
    [71]
    Moulin-Romsee G, D'hondt E, De Groot T, et al. Non-invasive grading of brain tumours using dynamic amino acid PET imaging: does it work for 11C-methionine?[J]. Eur J Nucl Med Mol Imaging, 2007, 34(12): 2082–2087. doi: 10.1007/s00259-007-0557-4
    [72]
    Kratochwil C, Combs SE, Leotta K, et al. Intra-individual comparison of 18F-FET and 18F-DOPA in PET imaging of recurrent brain tumors[J]. Neuro-Oncol, 2014, 16(3): 434–440. doi: 10.1093/neuonc/not199
    [73]
    Xiao J, Jin Y, Nie J, et al. Diagnostic and grading accuracy of 18F-FDOPA PET and PET/CT in patients with gliomas: a systematic review and meta-analysis[J]. BMC Cancer, 2019, 19(1): 767. doi: 10.1186/s12885-019-5938-0
    [74]
    Becherer A, Karanikas G, Szabó M, et al. Brain tumour imaging with PET: a comparison between [18F]fluorodopa and [11C]methionine[J]. Eur J Nucl Med Mol Imaging, 2003, 30(11): 1561–1567. doi: 10.1007/s00259-003-1259-1
    [75]
    Fueger BJ, Czernin J, Cloughesy T, et al. Correlation of 6–18F-fluoro-L-dopa PET uptake with proliferation and tumor grade in newly diagnosed and recurrent gliomas[J]. J Nucl Med, 2010, 51(10): 1532–1538. doi: 10.2967/jnumed.110.078592
    [76]
    Janvier L, Olivier P, Blonski M, et al. Correlation of SUV-derived indices with tumoral aggressiveness of gliomas in static 18F-FDOPA PET: use in clinical practice[J]. Clin Nucl Med, 2015, 40(9): e429–e435. doi: 10.1097/RLU.0000000000000897
    [77]
    Shields AF, Grierson JR, Dohmen BM, et al. Imaging proliferation in vivo with [F-18]FLT and positron emission tomography[J]. Nat Med, 1998, 4(11): 1334–1336. doi: 10.1038/3337
    [78]
    Hong IK, Kim JH, Ra YS, et al. Diagnostic usefulness of 3'-Deoxy-3'-[18F]Fluorothymidine positron emission tomography in recurrent brain tumor[J]. J Comput Assist Tomogr, 2011, 35(6): 679–684. doi: 10.1097/RCT.0b013e3182345b0e
    [79]
    Enslow MS, Zollinger LV, Morton KA, et al. Comparison of 18F-fluorodeoxyglucose and 18F-fluorothymidine PET in differentiating radiation necrosis from recurrent glioma[J]. Clin Nucl Med, 2012, 37(9): 854–861. doi: 10.1097/RLU.0b013e318262c76a
    [80]
    Shishido H, Kawai N, Miyake K, et al. Diagnostic value of 11C-methionine (MET) and 18F-fluorothymidine (FLT) positron emission tomography in recurrent high-grade gliomas; differentiation from treatment-induced tissue necrosis[J]. Cancers (Basel), 2012, 4(1): 244–256. doi: 10.3390/cancers4010244
    [81]
    Weber MA, Henze M, Tüttenberg J, et al. Biopsy targeting gliomas: do functional imaging techniques identify similar target areas?[J]. Invest Radiol, 2010, 45(12): 755–768. doi: 10.1097/RLI.0b013e3181ec9db0
    [82]
    Nowosielski M, Difranco MD, Putzer D, et al. An intra-individual comparison of MRI, [18F]-FET and [18F]-FLT PET in patients with high-grade gliomas[J]. PLoS One, 2014, 9(4): e95830. doi: 10.1371/journal.pone.0095830
    [83]
    Mertens K, Slaets D, Lambert B, et al. PET with 18F-labelled choline-based tracers for tumour imaging: a review of the literature[J]. Eur J Nucl Med Mol Imaging, 2010, 37(11): 2188–2193. doi: 10.1007/s00259-010-1496-z
    [84]
    Ito K, Yokoyama J, Kubota K, et al. Comparison of 18F-FDG and 11C-choline PET/CT for detecting recurrences in patients with nonsquamous cell head and neck malignancies[J]. Nucl Med Commun, 2010, 31(11): 931–937. doi: 10.1097/MNM.0b013e32833f3921
    [85]
    Kato T, Shinoda J, Nakayama N, et al. Metabolic assessment of gliomas using 11C-methionine, [18F] fluorodeoxyglucose, and 11C-choline positron-emission tomography[J]. AJNR Am J Neuroradiol, 2008, 29(6): 1176–1182. doi: 10.3174/ajnr.A1008
    [86]
    Hara T, Kondo T, Hara T, et al. Use of 18F-choline and 11C-choline as contrast agents in positron emission tomography imaging-guided stereotactic biopsy sampling of gliomas[J]. J Neurosurg, 2003, 99(3): 474–479. doi: 10.3171/jns.2003.99.3.0474
    [87]
    Montes A, Fernández A, Camacho V, et al. The usefulness of 18F-fluorocholine PET/CT in the detection of recurrence of central nervous system primary neoplasms[J]. Rev Esp Med Nucl Imagen Mol, 2017, 36(4): 227–232. doi: 10.1016/j.remn.2016.11.005
    [88]
    Gao L, Xu W, Li T, et al. Accuracy of 11C-choline positron emission tomography in differentiating glioma recurrence from radiation necrosis: a systematic review and meta-analysis[J]. Medicine (Baltimore), 2018, 97(29): e11556. doi: 10.1097/MD.0000000000011556
    [89]
    Dewhirst MW, Cao Y, Moeller B. Cycling hypoxia and free radicals regulate angiogenesis and radiotherapy response[J]. Nat Rev Cancer, 2008, 8(6): 425–437. doi: 10.1038/nrc2397
    [90]
    Gerstner ER, Zhang Z, Fink JR, et al. ACRIN 6684: assessment of tumor hypoxia in newly diagnosed glioblastoma using 18F-FMISO PET and MRI[J]. Clin Cancer Res, 2016, 22(20): 5079–5086. doi: 10.1158/1078-0432.CCR-15-2529
    [91]
    Reeves KM, Song PN, Angermeier A, et al. 18F-FMISO PET imaging identifies hypoxia and immunosuppressive tumor microenvironments and guides targeted evofosfamide therapy in tumors refractory to PD-1 and CTLA-4 inhibition[J]. Clin Cancer Res, 2022, 28(2): 327–337. doi: 10.1158/1078-0432.CCR-21-2394
    [92]
    Parent EE, Benayoun M, Ibeanu I, et al. [18F]Fluciclovine PET discrimination between high- and low-grade gliomas[J]. EJNMMI Res, 2018, 8(1): 67. doi: 10.1186/s13550-018-0415-3
    [93]
    Sörensen J, Owenius R, Lax M, et al. Regional distribution and kinetics of [18F]fluciclovine (anti-[18F]FACBC), a tracer of amino acid transport, in subjects with primary prostate cancer[J]. Eur J Nucl Med Mol Imaging, 2013, 40(3): 394–402. doi: 10.1007/s00259-012-2291-9
    [94]
    Kondo A, Ishii H, Aoki S, et al. Phase IIa clinical study of[18F]fluciclovine: efficacy and safety of a new PET tracer for brain tumors[J]. Ann Nucl Med, 2016, 30(9): 608–618. doi: 10.1007/s12149-016-1102-y
    [95]
    Wakabayashi T, Iuchi T, Tsuyuguchi N, et al. Diagnostic performance and safety of positron emission tomography using 18F-Fluciclovine in patients with clinically suspected high- or low-grade gliomas: a multicenter phase IIb trial[J]. Asia Ocean J Nucl Med Biol, 2017, 5(1): 10–21. doi: 10.22038/aojnmb.2016.7869
    [96]
    Wakabayashi T, Hirose Y, Miyake K, et al. Determining the extent of tumor resection at surgical planning with 18F-fluciclovine PET/CT in patients with suspected glioma: multicenter phase III trials[J]. Ann Nucl Med, 2021, 35(12): 1279–1292. doi: 10.1007/s12149-021-01670-z
    [97]
    Mashimo T, Pichumani K, Vemireddy V, et al. Acetate is a bioenergetic substrate for human glioblastoma and brain metastases[J]. Cell, 2014, 159(7): 1603–1614. doi: 10.1016/j.cell.2014.11.025
    [98]
    Yoshii Y, Furukawa T, Saga T, et al. Acetate/acetyl-CoA metabolism associated with cancer fatty acid synthesis: overview and application[J]. Cancer Lett, 2015, 356(2 Pt A): 211–216. doi: 10.1016/j.canlet.2014.02.019
    [99]
    Liu RS, Chang C, Chu LS, et al. PET imaging of brain astrocytoma with 1–11C-acetate[J]. Eur J Nucl Med Mol Imaging, 2006, 33(4): 420–427. doi: 10.1007/s00259-005-0023-0
    [100]
    Tsuchida T, Takeuchi H, Okazawa H, et al. Grading of brain glioma with 1–11C-acetate PET: comparison with 18F-FDG PET[J]. Nucl Med Biol, 2008, 35(2): 171–176. doi: 10.1016/j.nucmedbio.2007.11.004
    [101]
    Dienel GA, Popp D, Drew PD, et al. Preferential labeling of glial and meningial brain tumors with [2-14C]Acetate[J]. J Nucl Med, 2001, 42(8): 1243–1250. https://pubmed.ncbi.nlm.nih.gov/11483687/
    [102]
    Marik J, Ogasawara A, Martin-Mcnulty B, et al. PET of glial metabolism using 2–18F-fluoroacetate[J]. J Nucl Med, 2009, 50(6): 982–990. doi: 10.2967/jnumed.108.057356
    [103]
    Vassileva V, Braga M, Barnes C, et al. Effective detection and monitoring of glioma using [18F]FPIA PET imaging[J]. Biomedicines, 2021, 9(7): 811. doi: 10.3390/biomedicines9070811
    [104]
    Chin FT, Shen B, Liu S, et al. First experience with clinical-grade [18F]FPP(RGD)2: an automated multi-step radiosynthesis for clinical PET studies[J]. Mol Imaging Biol, 2012, 14(1): 88–95. doi: 10.1007/s11307-011-0477-3
    [105]
    Iagaru A, Mosci C, Mittra E, et al. Glioblastoma multiforme recurrence: an exploratory study of 18F FPPRGD2 PET/CT[J]. Radiology, 2015, 277(2): 497–506. doi: 10.1148/radiol.2015141550
    [106]
    Minamimoto R, Jamali M, Barkhodari A, et al. Biodistribution of the 18F-FPPRGD2 PET radiopharmaceutical in cancer patients: an atlas of SUV measurements[J]. Eur J Nucl Med Mol Imaging, 2015, 42(12): 1850–1858. doi: 10.1007/s00259-015-3096-4
    [107]
    Albert NL, Weller M, Suchorska B, et al. Response assessment in Neuro-oncology working group and European association for Neuro-oncology recommendations for the clinical use of PET imaging in gliomas[J]. Neuro Oncol, 2016, 18(9): 1199–1208. doi: 10.1093/neuonc/now058
    [108]
    Lin J, Chuang CP, Lin J, et al. Rational design, pharmacomodulation, and synthesis of [68Ga]Ga-Alb-FAPtp-01, a selective tumor-associated fibroblast activation protein tracer for PET imaging of Glioma[J]. ACS Sens, 2021, 6(9): 3424–3435. doi: 10.1021/acssensors.1c01316
    [109]
    Foster A, Nigam S, Tatum DS, et al. Novel theranostic agent for PET imaging and targeted radiopharmaceutical therapy of tumour-infiltrating immune cells in glioma[J]. EBioMedicine, 2021, 71: 103571. doi: 10.1016/j.ebiom.2021.103571
    [110]
    Alzheimer's Association. 2018 Alzheimer's disease facts and figures[J]. Alzheimer's Dement, 2018, 14(3): 367–429. doi: 10.1016/j.jalz.2018.02.001
    [111]
    Patterson C. World Alzheimer report 2018: the state of the art of dementia research: new frontiers[M]. London: Alzheimer's Disease International, 2018.
    [112]
    Breijyeh Z, Karaman R. Comprehensive review on Alzheimer's disease: causes and treatment[J]. Molecules, 2020, 25(24): 5789. doi: 10.3390/molecules25245789
    [113]
    Filippi L, Chiaravalloti A, Bagni O, et al. 18F-labeled radiopharmaceuticals for the molecular neuroimaging of amyloid plaques in Alzheimer's disease[J]. Am J Nucl Med Mol Imaging, 2018, 8(4): 268–281. https://pubmed.ncbi.nlm.nih.gov/30245918/
    [114]
    Beach TG. A review of biomarkers for neurodegenerative disease: will they swing us across the valley?[J]. Neurol Ther, 2017, 6(S1): 5–13. doi: 10.1007/s40120-017-0072-x
    [115]
    Gu L, Guo Z. Alzheimer's Aβ42 and Aβ40 peptides form interlaced amyloid fibrils[J]. J Neurochem, 2013, 126(3): 305–311. doi: 10.1111/jnc.12202
    [116]
    Kayed R, Lasagna-Reeves CA. Molecular mechanisms of amyloid oligomers toxicity[J]. J Alzheimers Dis, 2013, 33(S1): S67–S78. doi: 10.3233/JAD-2012-129001
    [117]
    Hu WT, Watts KD, Shaw LM, et al. CSF beta-amyloid 1–42-what are we measuring in Alzheimer's disease?[J]. Ann Clin Transl Neurol, 2015, 2(2): 131–139. doi: 10.1002/acn3.160
    [118]
    Uzuegbunam BC, Librizzi D, Hooshyar Yousefi B. PET radiopharmaceuticals for Alzheimer's disease and Parkinson's disease diagnosis, the current and future landscape[J]. Molecules, 2020, 25(4): 977. doi: 10.3390/molecules25040977
    [119]
    Klunk WE, Engler H, Nordberg A, et al. Imaging brain amyloid in Alzheimer's disease with Pittsburgh Compound-B[J]. Ann Neurol, 2004, 55(3): 306–319. doi: 10.1002/ana.20009
    [120]
    Forsberg A, Engler H, Almkvist O, et al. PET imaging of amyloid deposition in patients with mild cognitive impairment[J]. Neurobiol Aging, 2008, 29(10): 1456–1465. doi: 10.1016/j.neurobiolaging.2007.03.029
    [121]
    Okello A, Koivunen J, Edison P, et al. Conversion of amyloid positive and negative MCI to AD over 3 years: an 11C-PIB PET study[J]. Neurology, 2009, 73(10): 754–760. doi: 10.1212/WNL.0b013e3181b23564
    [122]
    Jack CR JR, Wiste HJ, Vemuri P, et al. Brain beta-amyloid measures and magnetic resonance imaging atrophy both predict time-to-progression from mild cognitive impairment to Alzheimer's disease[J]. Brain, 2010, 133(11): 3336–3348. doi: 10.1093/brain/awq277
    [123]
    Kemppainen NM, Scheinin NM, Koivunen J, et al. Five-year follow-up of 11C-PIB uptake in Alzheimer's disease and MCI[J]. Eur J Nucl Med Mol Imaging, 2014, 41(2): 283–289. doi: 10.1007/s00259-013-2562-0
    [124]
    Mathis CA, Ikonomovic MD, Debnath ML, et al. Comparison of the binding of 3′-F-PiB and PiB in human brain homogenates[J]. NeuroImage, 2008, 41(S2): T113–T114. https://www.proquest.com/docview/1506904887/fulltextPDF/E1CB33F6B4B14D3FPQ/1?accountid=42094
    [125]
    Vandenberghe R, Van Laere K, Ivanoiu A, et al. 18F-flutemetamol amyloid imaging in Alzheimer disease and mild cognitive impairment: a phase 2 trial[J]. Ann Neurol, 2010, 68(3): 319–329. doi: 10.1002/ana.22068
    [126]
    Juréus A, Swahn BM, Sandell J, et al. Characterization of AZD4694, a novel fluorinated Aβ plaque neuroimaging PET radioligand[J]. J Neurochem, 2010, 114(3): 784–794. doi: 10.1111/j.1471-4159.2010.06812.x
    [127]
    Verhoeff NPLG, Wilson AA, Takeshita S, et al. In-vivo imaging of Alzheimer disease beta-amyloid with[11C] SB-13 PET[J]. Am J Geriatr Psychiatry, 2004, 12(6): 584–595. doi: 10.1176/appi.ajgp.12.6.584
    [128]
    Johnson AE, Jeppsson F, Sandell J, et al. AZD2184: a radioligand for sensitive detection of β-amyloid deposits[J]. J Neurochem, 2009, 108(5): 1177–1186. doi: 10.1111/j.1471-4159.2008.05861.x
    [129]
    Nyberg S, Jönhagen ME, Cselényi Z, et al. Detection of amyloid in Alzheimer's disease with positron emission tomography using [11C]AZD2184[J]. Eur J Nucl Med Mol Imaging, 2009, 36(11): 1859–1863. doi: 10.1007/s00259-009-1182-1
    [130]
    Cselényi Z, Jönhagen ME, Forsberg A, et al. Clinical validation of 18F-AZD4694, an amyloid-β–specific PET radioligand[J]. J Nucl Med, 2012, 53(3): 415–424. doi: 10.2967/jnumed.111.094029
    [131]
    Rowe CC, Pejoska S, Mulligan RS, et al. Head-to-head comparison of 11C-PiB and 18F-AZD4694 (NAV4694) for β-amyloid imaging in aging and dementia[J]. J Nucl Med, 2013, 54(6): 880–886. doi: 10.2967/jnumed.112.114785
    [132]
    Giannakopoulos P, Herrmann FR, Bussière T, et al. Tangle and neuron numbers, but not amyloid load, predict cognitive status in Alzheimer's disease[J]. Neurology, 2003, 60(9): 1495–1500. doi: 10.1212/01.WNL.0000063311.58879.01
    [133]
    Van Rossum IA, Visser PJ, Knol DL, et al. Injury markers but not amyloid markers are associated with rapid progression from mild cognitive impairment to dementia in Alzheimer's disease[J]. J Alzheimers Dis, 2012, 29(2): 319–327. doi: 10.3233/JAD-2011-111694
    [134]
    Small GW, Kepe V, Ercoli LM, et al. PET of brain amyloid and tau in mild cognitive impairment[J]. N Engl J Med, 2006, 355(25): 2652–2663. doi: 10.1056/NEJMoa054625
    [135]
    Thompson PW, Ye L, Morgenstern JL, et al. Interaction of the amyloid imaging tracer FDDNP with hallmark Alzheimer's disease pathologies[J]. J Neurochem, 2009, 109(2): 623–630. doi: 10.1111/j.1471-4159.2009.05996.x
    [136]
    Okamura N, Suemoto T, Furumoto S, et al. Quinoline and benzimidazole derivatives: candidate probes for in vivo imaging of tau pathology in Alzheimer's disease[J]. J Neurosci, 2005, 25(47): 10857–1062. doi: 10.1523/JNEUROSCI.1738-05.2005
    [137]
    Fodero-Tavoletti MT, Okamura N, Furumoto S, et al. 18F-THK523: a novel in vivo tau imaging ligand for Alzheimer's disease[J]. Brain, 2011, 134(Pt 4): 1089–1100. doi: 10.1093/brain/awr038
    [138]
    Okamura N, Furumoto S, Harada R, et al. Novel 18F-labeled arylquinoline derivatives for noninvasive imaging of tau pathology in Alzheimer disease[J]. J Nucl Med, 2013, 54(8): 1420–1427. doi: 10.2967/jnumed.112.117341
    [139]
    Harada R, Okamura N, Furumoto S, et al. Characteristics of tau and its ligands in PET imaging[J]. Biomolecules, 2016, 6(1): 7. doi: 10.3390/biom6010007
    [140]
    Harada R, Okamura N, Furumoto S, et al. 18F-THK5351: a novel PET radiotracer for imaging neurofibrillary pathology in Alzheimer disease[J]. J Nucl Med, 2016, 57(2): 208–214. doi: 10.2967/jnumed.115.164848
    [141]
    Xia C, Arteaga J, Chen G, et al. [18F]T807, a novel tau positron emission tomography imaging agent for Alzheimer's disease[J]. Alzheimers Dement, 2013, 9(6): 666–676. doi: 10.1016/j.jalz.2012.11.008
    [142]
    Chhatwal JP, Schultz AP, Marshall GA, et al. Temporal T807 binding correlates with CSF tau and phospho-tau in normal elderly[J]. Neurology, 2016, 87(9): 920–926. doi: 10.1212/WNL.0000000000003050
    [143]
    Johnson KA, Schultz A, Betensky RA, et al. Tau positron emission tomographic imaging in aging and early Alzheimer disease[J]. Ann Neurol, 2016, 79(1): 110–119. doi: 10.1002/ana.24546
    [144]
    Vermeiren C, Motte P, Viot D, et al. The tau positron-emission tomography tracer AV-1451 binds with similar affinities to tau fibrils and monoamine oxidases[J]. Mov Disord, 2018, 33(2): 273–281. doi: 10.1002/mds.27271
    [145]
    Maruyama M, Shimada H, Suhara T, et al. Imaging of tau pathology in a tauopathy mouse model and in Alzheimer patients compared to normal controls[J]. Neuron, 2013, 79(6): 1094–1108. doi: 10.1016/j.neuron.2013.07.037
    [146]
    Kolb HC, Andrés JI. Tau positron emission tomography imaging[J]. Cold Spring Harb Perspect Biol, 2017, 9(5): a023721. doi: 10.1101/cshperspect.a023721
    [147]
    Sanabria Bohórquez S, Marik J, Ogasawara A, et al. [18F]GTP1 (Genentech Tau Probe 1), a radioligand for detecting neurofibrillary tangle tau pathology in Alzheimer's disease[J]. Eur J Nucl Med Mol Imaging, 2019, 46(10): 2077–2089. doi: 10.1007/s00259-019-04399-0
    [148]
    Wong DF, Comley RA, Kuwabara H, et al. Characterization of 3 novel tau radiopharmaceuticals, 11C-RO-963, 11C-RO-643, and 18F-RO-948, in healthy controls and in Alzheimer subjects[J]. J Nucl Med, 2018, 59(12): 1869–1876. doi: 10.2967/jnumed.118.209916
    [149]
    Kroth H, Oden F, Molette J, et al. Discovery and preclinical characterization of [18F]PI-2620, a next-generation tau PET tracer for the assessment of tau pathology in Alzheimer's disease and other tauopathies[J]. Eur J Nucl Med Mol Imaging, 2019, 46(10): 2178–2189. doi: 10.1007/s00259-019-04397-2
    [150]
    Shi Y, Murzin AG, Falcon B, et al. Cryo-EM structures of tau filaments from Alzheimer's disease with PET ligand APN-1607[J]. Acta Neuropathol, 2021, 141(5): 697–708. doi: 10.1007/s00401-021-02294-3
    [151]
    Hsu JL, Lin KJ, Hsiao IT, et al. The imaging features and clinical Associations of a novel tau PET tracer-18F-APN1607 in Alzheimer disease[J]. Clin Nucl Med, 2020, 45(10): 747–756. doi: 10.1097/RLU.0000000000003164
    [152]
    Walji AM, Hostetler ED, Selnick H, et al. Discovery of 6-(Fluoro-18F)-3-(1H-pyrrolo[2, 3-c]pyridin-1-yl)isoquinolin-5-amine ([18F]-MK-6240): A positron emission tomography (PET) imaging agent for quantification of neurofibrillary tangles (NFTs)[J]. J Med Chem, 2016, 59(10): 4778–4789. doi: 10.1021/acs.jmedchem.6b00166
    [153]
    Betthauser TJ, Cody KA, Zammit MD, et al. In vivo characterization and quantification of neurofibrillary tau PET Radioligand 18F-MK-6240 in humans from Alzheimer disease dementia to young controls[J]. J Nucl Med, 2019, 60(1): 93–99. doi: 10.2967/jnumed.118.209650
    [154]
    Rombouts FJR, Andrés JI, Ariza M, et al. Discovery of N-(Pyridin-4-yl)-1, 5-naphthyridin-2-amines as potential tau pathology PET tracers for Alzheimer's disease[J]. J Med Chem, 2017, 60(4): 1272–1291. doi: 10.1021/acs.jmedchem.6b01173
    [155]
    Declercq L, Rombouts F, Koole M, et al. Preclinical evaluation of 18F-JNJ64349311, a Novel PET tracer for tau imaging[J]. J Nucl Med, 2017, 58(6): 975–981. doi: 10.2967/jnumed.116.185199
    [156]
    Ehman EC, Johnson GB, Villanueva-Meyer JE, et al. PET/MRI: where might it replace PET/CT?[J]. J Magn Reson Imaging, 2017, 46(5): 1247–1262. doi: 10.1002/jmri.25711
    [157]
    Mainta IC, Vargas MI, Trombella S, et al. Hybrid PET-MRI in Alzheimer's disease research[M]//Perneczky R. Biomarkers for Alzheimer's Disease Drug Development. New York: Humana Press, 2018: 185–200.
    [158]
    Zhang M, Ni Y, Zhou Q, et al. 18F-florbetapir PET/MRI for quantitatively monitoring myelin loss and recovery in patients with multiple sclerosis: a longitudinal study[J]. eClinicalMedicine, 2021, 37: 100982. doi: 10.1016/j.eclinm.2021.100982
    [159]
    Zhang M, Sun W, Guan Z, et al. Simultaneous PET/fMRI detects distinctive alterations in functional connectivity and glucose metabolism of Precuneus Subregions in Alzheimer's disease[J]. Front Aging Neurosci, 2021, 13: 737002. doi: 10.3389/fnagi.2021.737002
    [160]
    Jabeen S, Arbind A, Kumar D, et al. Combined amino acid PET-MRI for identifying recurrence in post-treatment gliomas: together we grow[J]. Eur J Hybrid Imaging, 2021, 5(1): 15. doi: 10.1186/s41824-021-00109-y
    [161]
    Johannessen K, Berntsen EM, Johansen H, et al. 18F-FACBC PET/MRI in the evaluation of human brain metastases: a case report[J]. Eur J Hybrid Imaging, 2021, 5(1): 7. doi: 10.1186/s41824-021-00101-6
    [162]
    Bertaux M, Berenbaum A, Di Stefano AL, et al. Hybrid[18F]-F-DOPA PET/MRI interpretation criteria and scores for Glioma follow-up after radiotherapy[J]. Clin Neuroradiol, 2022, 32(3): 735–747. doi: 10.1007/s00062-022-01139-0
  • Cited by

    Periodical cited type(2)

    1. Kemter AM, Patry RT, Arnold J, et al. Commensal bacteria signal through TLR5 and AhR to improve barrier integrity and prevent allergic responses to food. Cell Rep, 2023, 42(10): 113153. DOI:10.1016/j.celrep.2023.113153
    2. Liu J, Guo C, Wang Y, et al. Preclinical insights into fucoidan as a nutraceutical compound against perfluorooctanoic acid-associated obesity via targeting endoplasmic reticulum stress. Front Nutr, 2022, 9: 950130. DOI:10.3389/fnut.2022.950130

    Other cited types(0)

Catalog

    Tables(3)

    Article Metrics

    Article views (1094) PDF downloads (188) Cited by(2)
    Related

    /

    DownLoad:  Full-Size Img  PowerPoint
    Return
    Return