Abstract
The immune system’s role in atherosclerosis has long been an important research topic and is increasingly investigated for therapeutic and diagnostic purposes. Therefore, noninvasive imaging of hematopoietic organs and immune cells will undoubtedly improve atherosclerosis phenotyping and serve as a monitoring method for immunotherapeutic treatments. Among the available imaging techniques, positron emission tomography (PET)’s unique features make it an ideal tool to quantitatively image the immune response in the context of atherosclerosis and afford reliable readouts to guide medical interventions in cardiovascular disease. Here, we summarize the state of the art in the field of atherosclerosis PET immunoimaging and provide an outlook on current and future applications.
Introduction
Despite recent advances in its management, cardiovascular disease (CVD) continues to be the leading cause of mortality worldwide. CVD’s main underlying cause, atherosclerosis, is a slowly progressing, chronic disease of the arterial wall. While originally the focus has been on lipids, mounting evidence accumulated over the past 25 years has unequivocally established inflammation as a key driver of atherogenesis. The disease process is initiated by endothelium disruption and the accumulation of oxidized low-density lipoprotein (oxLDL), which triggers immune cell recruitment to the vessel wall. In early lesions, monocytes differentiate into macrophages, which upon oxLDL uptake evolve into foam cells. Further accumulation of lipids leads to foam cell death and vessel wall thickening. These focal lesions, called plaques, can eventually rupture and the released material trigger acute cardiovascular events such as myocardial infarction or stroke. Therefore, unraveling the immune system’s role in atherosclerosis is expected to increasingly lead to viable immunotherapeutic interventions, while – similar to cancer immunotherapy – patient stratification and treatment monitoring can be facilitated by imaging approaches.
Indeed, two recent clinical trials highlight the complex role that inflammation plays in atherosclerotic CVD. The CANTOS trial concluded that anti-interleukin 1β therapy results in a reduced rate of recurrent cardiovascular events.1 On the other hand, the CIRT study demonstrated that low dose methotrexate treatment did not result in lower levels of a number of inflammatory markers nor in fewer cardiovascular events compared to placebo.2 These mixed results likely indicate the existence of specific inflammatory pathways relevant to atherosclerosis, but also prove that effectively targeting the involved pathways systemically is a plausible therapeutic avenue to treat CVD. Consequently, atherosclerosis immunology’s complexity and the resulting arterial wall remodeling mandate an integrative approach probing the hematopoietic system as well as plaque activity, morphology and composition.
Positron emission tomography (PET) is particularly suited for this purpose due to this imaging modality’s high tissue penetration and sensitivity for the detection of radiopharmaceuticals with specific biological targets. PET allows noninvasive quantification of molecular processes in a hot spot fashion using minimal amounts of injected radiotracer with no pharmacological effects. While PET necessitates anatomical reference and poses radiation concerns, its value for atherosclerosis imaging – in combination with computed tomography (CT) or magnetic resonance imaging (MRI) – is increasingly appreciated. For example, a recent study by Sanz and colleagues has shown PET/MRI’s potential to detect subclinical atherosclerosis.3 The advent of total-body PET is expected to allow simultaneous vessel wall and hematopoietic system imaging, at reduced scan times and, most importantly, at a relatively low radiotracer dose due to its substantially increased sensitivity.4 The latter is simultaneously facilitated by advances in probe design, consisting of molecular ligands – ranging from small molecules to complex proteins – and their labeling with radioisotopes, which can be achieved by covalent conjugation or through chelation.
In this review, we summarize the growing body of work focused on PET imaging of the immune system in atherosclerosis, highlighting its value as a tool in anti-atherosclerosis drug development, and to diagnose and guide therapeutic interventions.
Imaging the immune response in atherosclerosis by PET
Atherosclerosis PET imaging initially focused on the so-called vulnerable plaque, aiming to identify rupture-prone lesions likely to trigger acute events. Immunologically, an increasing number of cell types is being implicated in atheroprogression5 but monocytes/macrophages remain by far the preferred cellular target for immunoimaging of atherosclerosis given their prominent role in plaque development and rupture. While identifying high-risk plaques is still a key goal, the paradigm shift in atherosclerosis research towards a more integrative systems approach is also slowly changing how the disease is imaged. It has long been known that high numbers of circulating leukocytes are predictive of cardiovascular events.6 In mice, this monocytosis is also observed,7,8 and results from increased hematopoietic activity.9 Moreover, CVD events like myocardial infarction aggravate atherosclerosis through heightened hematopoiesis and the ensuing monocytosis.10 These findings have been corroborated non-invasively by PET imaging. Increased hematopoiesis has been measured as augmented proliferative/metabolic activity in the spleen and bone marrow in different animal models9 as well as in humans11 with atherosclerosis. Moreover, in another PET imaging-based study, psychosocial stress has been linked to CVD events through an association between resting amygdalar activity and medullary hematopoiesis.12 Collectively, these findings support the role of the hematopoietic system in the low-grade inflammation observed in atherosclerosis, as well as the need to assess the disease systemically.
Many radiotracers that were initially developed for oncological PET have found application in atherosclerosis immunoimaging, including fluorine-18 (18F)-labeled 2-deoxy-2-fluoro-D-glucose (18F-FDG) and 3’-deoxy-3’-fluorothymidine (18F-FLT) or gallium-68 (68Ga)-labeled DOTATATE. While not directly targeting immune cells or the immune function, it is worth mentioning 18F-sodium fluoride (18F-NaF) as an atherosclerosis PET radiotracer. 18F-NaF accumulates in plaques through its binding to calcium phosphate (mostly hydroxyapatite) in microcalcifications,13 which are produced by smooth muscle cell-derived osteoblast-like cells in response to inflammatory cytokines. Given its fast clearance and very low myocardial uptake, 18F-NaF can efficiently visualize high-risk and ruptured atherosclerotic plaques in the coronary arteries.14 However, 18F-NaF atherosclerosis PET imaging is not devoid of limitations, as its high accumulation in the bone may interfere with quantification of aortic microcalcifications.
Among the available PET radioisotopes, 18F is the most widely used for labeling of small molecules, while zirconium-89 (89Zr) and the generator-produced gallium-68 (68Ga) are gaining traction for labeling peptides, proteins and nanoparticles through chelation. Table 1 shows the most relevant radionuclides used for atherosclerosis immunoimaging and some key physicochemical properties. Figure 1 schematically summarizes the main atherosclerosis PET immunoimaging approaches that will be discussed in the following sections, as well as a selection of probes and labeling strategies thus far implemented.
Table 1.
Radioisotopes most frequently used for atherosclerosis immunoimaging by positron emission tomography. t1/2 = physical half-life; Rmean = mean positron range; EC = electron capture; [C] = cyclotron; [G] = generator.
| Isotope | t1/2 [h] | Production | Decay | Rmean [mm] | Probe | References |
|---|---|---|---|---|---|---|
| 11C | 0.34 | 14N(p,α)11C [C] | >99% β+ | 1.27 |
11C-choline 11C-acetate 11C-PK11195 |
27 29 40,41 |
| 18F | 1.83 | 18O(p,n)18F [C] | 96.9% β+ 3.1% EC |
0.66 |
18F-FDG 18F-FDM 18F-FLT 18F-choline 18F-macroflor |
3,11,12,18–20 25 9,60 28 58 |
| 68Ga | 1.13 | 68Ge/68Ga [G] | 89.1% β+ 10.9% EC |
3.56 |
68Ga-DOTATATE 68Ga-DOTATOC 68Ga-nanobodies |
30,31 33,34 46,50 |
| 64Cu | 12.7 | 64Ni(p,n)64Cu [C] | 17.9% β+ 38.4% β− 43.1% EC |
0.56 |
64Cu-DOTATATE 64Cu-nanobodies |
32,33 50 |
| 89Zr | 78.4 | 89Y(p,n)89Zr [C] | 22.3% β+ 76.6% EC |
1.27 |
89Zr-HDL 89Zr-hyaluronan 89Zr-LA25 |
57 59 52 |
Figure 1. Atherosclerosis immunoimaging by positron emission tomography.

A) PET imaging allows both focal and systems imaging of the atherosclerotic process by quantitative assessment of immune activation in the plaque and other tissues. Sp: spleen; BM: bone marrow. B) Key strategies for immunoimaging of atherosclerosis implemented to date. C) Selected PET radiotracers and radiolabeling strategies used for atherosclerosis PET immunoimaging. Chelators are shown in blue. NOTA: 1,4,7-Triazacyclononane-1,4,7-triacetic acid. DFO: deferoxamine.
Immunometabolic PET imaging
Activated immune cells show a distinct metabolic profile in contrast with quiescent states.15 Similar to tumor cells, immune cells undergo metabolic reprogramming towards aerobic glycolysis upon activation by external stimuli.16 Importantly, activation of glycolytic pathways occurs both in classically and alternatively activated macrophages.17 This metabolic shift reflects the energetic and biosynthetic demands of the activated cell. Thus, most metabolic radiotracers developed for cancer PET imaging have been used for atherosclerosis imaging. Among these, the glucose analog 18F-FDG18,19 features prominently. 18F-FDG is the most widely used radiotracer in the clinic accounting for 90% of oncological PET scans worldwide, and is also the preferred radiotracer for atherosclerosis imaging.20 In the plaque, 18F-FDG uptake correlates with macrophage burden and is used as an inflammation radiotracer,19 although enhanced 18F-FDG uptake may also be due to hypoxia.21 Despite its widespread use, 18F-FDG suffers from lack of specificity22 and high brain and myocardial uptake, which precludes imaging of the vasculature in these tissues. However, specific patient preparation protocols23,24 based on a ketogenic diet and fasting prior to the scan can efficiently suppress myocardial 18F-FDG uptake in most subjects and facilitate coronary artery wall imaging. As an alternative metabolic tracer to 18F-FDG, the use of 18F-labeled 2-deoxy-2-fluoro-D-mannose (18F-FDM) was explored in rabbits. In this study, both epimeric tracers were compared head to head showing very similar performance including myocardial uptake.25
At the same time, activated immune cells enter a proliferative state16 that requires biosynthetic components and building blocks including nucleotides. A study in mice, rabbits and humans showed that the uptake of 18F-labeled thymidine (18F-FLT)9 was significantly higher in subjects with atherosclerosis compared to controls due to plaque macrophage proliferation26. Moreover, 18F-FLT-PET in mice also revealed increased uptake in bone marrow and spleen, indicative of systemic immune activation in atherosclerosis. Similarly, proliferating macrophages need to synthesize membrane components like cholesterol, fatty acids or phospholipids.16 Exploiting this metabolic feature, 11C- and 18F-labeled choline, a building block for membrane phospholipids, have been used for imaging inflammation in mice27 and patients,28 respectively, showing increased uptake in plaques that was mainly due to macrophages. Along the same line, the use of 11C-acetate, which feeds fatty acid synthesis pathways, has been explored in patients.29 Interestingly, most calcified regions did not show enhanced 11C-acetate uptake, suggesting that macrophage activation and calcification are separate events in time.
Immune cell radioligand/receptor-based PET imaging
The abovementioned 18F-FDG’s shortcomings for atherosclerosis imaging, shared in part by all metabolic tracers, have spurred a search for higher specificity tracers and the ability to image the coronaries. A prevalent strategy focuses on targeting membrane receptors that are specifically upregulated in activated immune cells. One such example is the somatostatin receptor subtype 2 (SSR2), which is expressed on activated macrophages. A derivative of the somatostatin analog octreotide modified with the chelator DOTA, namely DOTA-octreotate or DOTATATE, has been extensively studied in the context of atherosclerosis imaging. Thus far, gallium-68 (68Ga)-labeled DOTATATE has been evaluated in mice, in comparison with other SSR2 tracers,30 and in patients, showing excellent macrophage specificity and superior coronary lesion discriminating features compared to 18F-FDG.31 The use of copper-64 (64Cu) instead of 68Ga allows imaging at later time points with improved spatial resolution – due to 64Cu shorter positron range – albeit potentially increasing radiation exposure. Using PET /MRI, 64Cu-DOTATATE was found to accumulate in carotid plaques, and this uptake correlated with alternatively activated macrophage burden.32 In a retrospective study in oncological patients, 64Cu-DOTATATE uptake correlated with CVD risk factors.33 Similarly, DOTATOC, another DOTA-modified octreotide peptide, has also been explored as a macrophage imaging agent in atherosclerosis in its 68Ga-labeled version with opposing results. In a head-to-head comparison with 64Cu-DOTATATE, 68Ga-DOTATOC uptake was significantly lower in vascular regions and did not correlate with CVD risk factors.33 A more recent study, on the contrary, reported a significant association between 68Ga-DOTATOC thoracic aortic uptake and cardiovascular risk factors.34
Similarly, the 68Ga-labeled CXC-motif chemokine receptor 4 (CXCR4) ligand pentixafor has been proposed as an inflammation radiotracer in atherosclerosis. While CXCR4 is expressed on several immune cell subsets, in the context of atherosclerosis it is found mainly on monocytes and macrophages.35 68Ga-pentixafor was first evaluated in rabbits36 with promising results, demonstrating uptake in macrophage-rich areas. More recently, two retrospective studies in patients established a significant correlation between 68Ga-pentixafor artery wall uptake and a number of CVD risk factors.37,38 Also in humans, 68Ga-pentixafor has been used to detect coronary artery wall inflammation by motion-corrected PET/CT.39 Collectively, these results indicate the clinical value of 68Ga-pentixafor as a macrophage-specific radiotracer to quantify arterial inflammation.
Translocator protein (TSPO) expression is upregulated in activated macrophages, representing another potential target to identify inflamed atherosclerotic plaques. To date, several TSPO ligands have been explored as atherosclerosis PET imaging agents. 11C-PK11195 PET imaging was first described in mice, showing increased accumulation in atherosclerotic plaques but with high non-specific uptake.40 In humans, 11C-PK11195 uptake was higher in carotid plaques associated with ipsilateral symptoms (stroke or transient ischemia) compared to those without.41 In addition, the TSPO-targeted radioligands 18F-FEMPA42, 18F-GE-18043 and 18F-PBR11144 have been tested in mice. While 18F-FEMPA and 18F-GE-180 showed specific uptake in macrophage-rich areas, their overall aortic uptake in atherosclerotic samples was not different from healthy tissue. On the other hand, 18F-PBR111 uptake was significantly higher in atherosclerotic aortas, where TSPO localized to CD11b+ monocyte-derived macrophages.44
Immuno-PET for atherosclerosis imaging
Immuno-PET, i.e. the use of antibody-based radiotracers for PET imaging, is an established approach in oncology.45 Due to their high affinity and target specificity, antibodies are ideal for imaging purposes. However, the use of full-size antibodies is undesirable in the context of atherosclerosis due to blood pool signal and spillover to the arterial wall as a result of their very long circulation time. For this reason, smaller antibody fragment-based radiotracers for PET and single-photon emission computed tomography (SPECT) have been developed and tested in mice, such as nanobodies targeting the macrophage mannose receptor (MMR),46 lectin-like oxidized LDL receptor-1 (LOX-1),47 and vascular cell adhesion molecule (VCAM)-1,48,49 an early marker of endothelium activation and immune cell recruitment. Nanobodies’ fast renal clearance facilitates imaging of the vessel wall due to reduced blood pool background signal, and affords highly specific imaging of their molecular targets.
More recently, we have used translational PET/MR imaging to study these three nanobodies in a head-to-head comparison using a rabbit atherosclerosis model.50 Initially labeled with 64Cu, the nanobodies were also compared with 18F-FDG. Further, the MMR nanobody was labeled with 68Ga and included in an atherosclerosis progression study that also comprised 18F-FDG- and 18F-NaF-PET imaging, in addition to MR-based assessment of vessel wall thickness and permeability. Interestingly, 68Ga-MMR nanobody uptake correlated with vessel wall area, demonstrating macrophage accumulation during atherosclerosis progression.50
The atherosclerotic process can also be monitored indirectly through evaluation of oxidative stress. It is becoming increasingly clear that oxidized phospholipids and other lipid peroxidation breakdown products are causative of CVD.51 In a recent study, we reported the development of an antigen-binding fragment (Fab)-based 89Zr-labeled radiotracer, derived from a natural antibody, that targets the malondialdehyde-acetaldehyde adduct oxidation-specific epitope and enables non-invasive visualization of atherosclerotic lesions.52 The Fab radiotracer uptake in the plaque correlated with 18F-FDG and with macrophage burden.
Immuno-PET imaging of atherosclerosis using small antibody fragments is therefore a compelling tool to non-invasively phenotype immune cells in vivo.53,54 This information, obtained focally in atherosclerotic lesions or systemically in hematopoietic tissues, can potentially be used to stratify patients for targeted therapies and/or to monitor response to treatment.54 Due to their fast clearance, one key strength of this approach is that it allows radiolabeling with short-lived isotopes and same-day imaging, which makes it more convenient for patients and reduces overall radiation exposure.55 On the other hand, antibody fragment development and production is costly, and their fast clearance has the downside of producing lower signals compared to full-size antibodies. However, this issue can be mitigated through chemical modification in order to increase their size and thus prolong their circulation times.54
Phagocyte-mediated nanoparticle uptake PET imaging
Lastly, radiolabeled nanoparticles can be used for atherosclerosis immunoimaging by PET. High-density lipoprotein (HDL), a natural nanoparticle, has an intrinsic ability to interact with macrophages through membrane receptors in reverse cholesterol transport, a feature that has been exploited for therapeutic and imaging purposes in the past.56 More recently, we developed 89Zr radiolabeling strategies that enabled non-invasive visualization of HDL in mouse, rabbit and swine atherosclerosis models, showing specific uptake by macrophages.57 Importantly, these radiolabeling strategies have been subsequently used for characterization of HDL-based nanobiologics.56 However, HDL’s long circulation time prohibits its use as an atherosclerosis imaging agent. Conversely, an 18F-labeled macrophage-targeted polyglucose nanoparticle, named 18F-Macroflor, has been evaluated in mice and rabbits.58 18F-Macroflor’s small size (~5 nm) allows fast renal clearance and low background signal for vessel wall imaging. This nanotracer is taken up by macrophages, and was evaluated not only for atherosclerosis imaging, but also for myocardial infarction in mice. Similarly, we radiolabeled a hyaluronan nanoparticle with 89Zr to image macrophage inflammation in atherosclerotic mice and rabbits.59
Perspective/conclusion
Atherosclerosis immunoimaging by PET is a valuable approach to quantitatively assess disease burden, focally and systemically. Preclinically, it can be used to provide reliable quantitative efficacy data in early drug development. Thus, we advocate the integration of specific PET imaging readouts into multiparametric and translational PET/MR imaging protocols for evaluation of novel anti-atherosclerosis therapies in large animal models. In a recent study, we implemented this approach to escalate a nanoimmunotherapy from murine to rabbit and swine atherosclerosis models, which allowed us to assess treatment response from different angles using restricted group sizes (Figure 2).60 In clinical trials, immunoimaging by PET can be used to interrogate specific disease aspects and obtain surrogate imaging end points to assess treatment response without the need for large cohort numbers or long follow-up periods.61 While genetic background is an important disease risk determinant, phenotypical traits are more strongly correlated with disease progression and outcome. Ultimately, clinical PET immunoimaging of atherosclerosis may be used to noninvasively quantify specific disease markers, which combined with gene screening and/or liquid biopsy biomarker data may be used to tailor personalized interventions, or to guide patient selection for immunotherapy and its subsequent monitoring. In summary, we anticipate that atherosclerosis PET immunoimaging will mature into a powerful platform that can be integrated in early drug development and in CVD precision medicine to inform personalized treatment options.
Figure 2. Integration of PET atherosclerosis immunoimaging in translational drug development.

Imaging immune cell activity in the vessel wall can be integrated into anti-atherosclerosis (nano)therapy development to provide valuable information on treatment response. A) In a recent study using a swine model, we measured the response to a 2-week simvastatin high-density lipoprotein (S-HDL) nanotherapeutic regimen using a longitudinal PET/MR imaging protocol. B) This protocol combined 18F-FDG and 18F-FLT PET immunoimaging to measure vessel wall inflammation and macrophage proliferation (top), respectively, with MRI-derived vessel wall permeability and plaque burden measurements (bottom). This approach allowed us to obtain meaningful results on the therapy’s effect on overall disease burden using restricted group sizes. Adapted from Binderup et al60 with permission. Copyright © 2019, The American Association for the Advancement of Science.
Supplementary Material
Highlights.
The immune system is critically involved in atherosclerosis progression
PET is a quantitative imaging technique that allows noninvasive assessment of the immune system in atherosclerosis
Atherosclerosis PET immunoimaging has been achieved using metabolic tracers, radioligands for specific receptors, nanoparticles and small antibody fragments targeting immune cell markers
PET immunoimaging can be used to phenotype atherosclerotic plaques, assess hematopoietic tissue activation or guide CVD treatment
Sources of Funding:
This work was supported by the National Institutes of Health grants R01HL071021, R01HL128056, R01HL144072, R01HL135878, R01HL143814, P01 HL131478 (Z.A.F.); R01 HL125703, R01 HL118440 (W.J.M.M.); the American Heart Association 16SDG31390007 (C.P.M.); and the Netherlands Organization for Scientific Research NWO Vidi (W.J.M.M).
Nonstandard abbreviations and acronyms
- PET
positron emission tomography
- CVD
cardiovascular disease
- CT
computed tomography
- MRI
magnetic resonance imaging
- FDG
fluorodeoxyglucose
- DOTA
1,4,7,10-tetraazacyclododecane-1,4,7,10-tetraacetic acid
- HDL
high-density lipoprotein
- oxLDL
oxidized low-density lipoprotein
Footnotes
Disclosure: The authors declare no conflict of interest.
References
- 1.Ridker PM, Everett BM, Thuren T, et al. Antiinflammatory Therapy with Canakinumab for Atherosclerotic Disease. N Engl J Med. 2017;377:1119–1131. doi: 10.1056/NEJMoa1707914. [DOI] [PubMed] [Google Scholar]
- 2.Ridker PM, Everett BM, Pradhan A, et al. Low-Dose Methotrexate for the Prevention of Atherosclerotic Events. N Engl J Med. 2019;380:752–762. doi: 10.1056/NEJMoa1809798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Fernández-Friera L, Fuster V, López-Melgar B, et al. Vascular Inflammation in Subclinical Atherosclerosis Detected by Hybrid PET/MRI. J Am Coll Cardiol. 2019;73:1371–1382. doi: 10.1016/J.JACC.2018.12.075. [DOI] [PubMed] [Google Scholar]
- 4.Badawi RD, Shi H, Hu P, Chen S, Xu T, Price PM, Ding Y, Spencer BA, Nardo L, Liu W, Bao J, Jones T, Li H, Cherry SR. First Human Imaging Studies with the EXPLORER Total-Body PET Scanner. J Nucl Med. 2019;60:299–303. doi: 10.2967/jnumed.119.226498. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Zhao TX, Mallat Z. Targeting the Immune System in Atherosclerosis: JACC State-of-the-Art Review. J Am Coll Cardiol. 2019;73:1691–1706. doi: 10.1016/J.JACC.2018.12.083. [DOI] [PubMed] [Google Scholar]
- 6.Friedman GD, Klatsky AL, Siegelaub AB. The Leukocyte Count as a Predictor of Myocardial Infarction. N Engl J Med. 1974;290:1275–1278. doi: 10.1056/NEJM197406062902302. [DOI] [PubMed] [Google Scholar]
- 7.Swirski FK, Libby P, Aikawa E, Alcaide P, Luscinskas FW, Weissleder R, Pittet MJ. Ly-6Chi monocytes dominate hypercholesterolemia-associated monocytosis and give rise to macrophages in atheromata. J Clin Invest. 2007;117:195–205. doi: 10.1172/JCI29950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Du W, Wong C, Song Y, Shen H, Mori D, Rotllan N, Price N, Dobrian AD, Meng H, Kleinstein SH, Fernandez-Hernando C, Goldstein DR. Age-associated vascular inflammation promotes monocytosis during atherogenesis. Aging Cell. 2016;15:766–777. doi: 10.1111/acel.12488. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Ye Y-X, Calcagno C, Binderup T, et al. Imaging Macrophage and Hematopoietic Progenitor Proliferation in Atherosclerosis. Circ Res. 2015;117;835–845. doi: 10.1161/CIRCRESAHA.115.307024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Dutta P, Courties G, Wei Y, et al. Myocardial infarction accelerates atherosclerosis. Nature. 2012;487:325–329. doi: 10.1038/nature11260. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.van der Valk FM, Kuijk C, Verweij SL, Stiekema LCA, Kaiser Y, Zeerleder S, Nahrendorf M, Voermans C, Stroes ESG. Increased haematopoietic activity in patients with atherosclerosis. Eur Heart J. 2016;38:ehw246. doi: 10.1093/eurheartj/ehw246. [DOI] [PubMed] [Google Scholar]
- 12.Tawakol A, Ishai A, Takx RA, et al. Relation between resting amygdalar activity and cardiovascular events: a longitudinal and cohort study. Lancet. 2017;389:834–845. doi: 10.1016/S0140-6736(16)31714-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Irkle A, Vesey AT, Lewis DY, Skepper JN, Bird JLE, Dweck MR, Joshi FR, Gallagher F a., Warburton E a., Bennett MR, Brindle KM, Newby DE, Rudd JH, Davenport AP. Identifying active vascular microcalcification by 18F-sodium fluoride positron emission tomography. Nat Commun. 2015;6:7495. doi: 10.1038/ncomms8495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Joshi NV, Vesey AT, Williams MC, et al. 18F-fluoride positron emission tomography for identification of ruptured and high-risk coronary atherosclerotic plaques: a prospective clinical trial. Lancet. 2014;383:705–713. doi: 10.1016/S0140-6736(13)61754-7. [DOI] [PubMed] [Google Scholar]
- 15.Pearce EL, Pearce EJ. Metabolic Pathways in Immune Cell Activation and Quiescence. Immunity. 2013;38:633–643. doi: 10.1016/J.IMMUNI.2013.04.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Koelwyn GJ, Corr EM, Erbay E, Moore KJ. Regulation of macrophage immunometabolism in atherosclerosis. Nat Immunol. 2018;19:526–537. doi: 10.1038/s41590-018-0113-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Tavakoli S, Short JD, Downs K, Nguyen HN, Lai Y, Zhang W, Jerabek P, Goins B, Sadeghi MM, Asmis R. Differential Regulation of Macrophage Glucose Metabolism by Macrophage Colony-stimulating Factor and Granulocyte-Macrophage Colony-stimulating Factor: Implications for 18F FDG PET Imaging of Vessel Wall Inflammation. Radiology. 2016:160839. doi: 10.1148/radiol.2016160839. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Rudd JHF, Warburton EA, Fryer TD, Jones HA, Clark JC, Antoun N, Johnströ P, Davenport AP, Kirkpatrick PJ, Arch BN, Pickard JD, Weissberg PL. Imaging Atherosclerotic Plaque Inflammation With [18F]-Fluorodeoxyglucose Positron Emission Tomography. Circulation. 2002;105:2708–2711. doi: 10.1161/01.CIR.0000020548.60110.76. [DOI] [PubMed] [Google Scholar]
- 19.Tawakol A, Migrino RQ, Bashian GG, Bedri S, Vermylen D, Cury RC, Yates D, LaMuraglia GM, Furie K, Houser S, Gewirtz H, Muller JE, Brady TJ, Fischman AJ. In vivo 18F-fluorodeoxyglucose positron emission tomography imaging provides a noninvasive measure of carotid plaque inflammation in patients. J Am Coll Cardiol. 2006;48:1818–1824. doi: 10.1016/j.jacc.2006.05.076. [DOI] [PubMed] [Google Scholar]
- 20.McKenney-Drake ML, Moghbel MC, Paydary K, Alloosh M, Houshmand S, Moe S, Salavati A, Sturek JM, Territo PR, Weaver C, Werner TJ, Høilund-Carlsen PF, Sturek M, Alavi A. 18F-NaF and 18F-FDG as molecular probes in the evaluation of atherosclerosis. Eur J Nucl Med Mol Imaging. 2018;45:2190–2200. doi: 10.1007/s00259-018-4078-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Folco EJ, Sheikine Y, Rocha VZ, Christen T, Shvartz E, Sukhova GK, Di Carli MF, Libby P. Hypoxia But Not Inflammation Augments Glucose Uptake in Human Macrophages. J Am Coll Cardiol. 2011;58:603–614. doi: 10.1016/j.jacc.2011.03.044. [DOI] [PubMed] [Google Scholar]
- 22.Iwaki T, Mizuma H, Hokamura K, Onoe H, Umemura K. [ 18 F]FDG Uptake in the Aortic Wall Smooth Muscle of Atherosclerotic Plaques in the Simian Atherosclerosis Model. Biomed Res Int. 2016;2016:1–12. doi: 10.1155/2016/8609274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Demeure F, Hanin F-X, Bol A, Vincent M-F, Pouleur A-C, Gerber B, Pasquet A, Jamar F, Vanoverschelde J-LJ, Vancraeynest D. A randomized trial on the optimization of 18F-FDG myocardial uptake suppression: implications for vulnerable coronary plaque imaging. J Nucl Med. 2014;55(10):1629–1635. doi: 10.2967/jnumed.114.138594. [DOI] [PubMed] [Google Scholar]
- 24.Christopoulos G, Jouni H, Acharya GA, Blauwet LA, Kapa S, Bois J, Chareonthaitawee P, Rodriguez-Porcel MG. Suppressing physiologic 18-fluorodeoxyglucose uptake in patients undergoing positron emission tomography for cardiac sarcoidosis: The effect of a structured patient preparation protocol. J Nucl Cardiol. 2019:1–11. doi: 10.1007/s12350-019-01746-4. [DOI] [PubMed] [Google Scholar]
- 25.Tahara N, Mukherjee J, de Haas HJ, et al. 2-deoxy-2-[18F]fluoro-d-mannose positron emission tomography imaging in atherosclerosis. Nat Med. 2014;20:215–219. doi: 10.1038/nm.3437. [DOI] [PubMed] [Google Scholar]
- 26.Robbins CS, Hilgendorf I, Weber GF, et al. Local proliferation dominates lesional macrophage accumulation in atherosclerosis. Nat Med. 2013;19:1166–1172. doi: 10.1038/nm.3258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Laitinen IEK, Luoto P, Någren K, Marjamäki PM, Silvola JMU, Hellberg S, Laine VJO, Ylä-Herttuala S, Knuuti J, Roivainen A. Uptake of 11C-choline in mouse atherosclerotic plaques. J Nucl Med. 2010;51:798–802. doi: 10.2967/jnumed.109.071704. [DOI] [PubMed] [Google Scholar]
- 28.Vöö S, Kwee RM, Sluimer JC, Schreuder FHBM, Wierts R, Bauwens M, Heeneman S, Cleutjens JPM, van Oostenbrugge RJ, Daemen J-WH, Daemen MJAP, Mottaghy FM, Kooi ME. Imaging Intraplaque Inflammation in Carotid Atherosclerosis With 18 F-Fluorocholine Positron Emission Tomography–Computed Tomography. Circ Cardiovasc Imaging. 2016;9. doi: 10.1161/CIRCIMAGING.115.004467. [DOI] [PubMed] [Google Scholar]
- 29.Derlin T, Habermann CR, Lengyel Z, Busch JD, Wisotzki C, Mester J, Pávics L. Feasibility of 11C-acetate PET/CT for imaging of fatty acid synthesis in the atherosclerotic vessel wall. J Nucl Med. 2011;52:1848–1854. doi: 10.2967/jnumed.111.095869. [DOI] [PubMed] [Google Scholar]
- 30.Rinne P, Hellberg S, Kiugel M, et al. Comparison of Somatostatin Receptor 2-Targeting PET Tracers in the Detection of Mouse Atherosclerotic Plaques. Mol Imaging Biol. 2016;18:99–108. doi: 10.1007/s11307-015-0873-1. [DOI] [PubMed] [Google Scholar]
- 31.Tarkin JM, Joshi FR, Evans NR, et al. Detection of Atherosclerotic Inflammation by 68Ga-DOTATATE PET Compared to [18F]FDG PET Imaging. J Am Coll Cardiol. 2017;69:1774–1791. doi: 10.1016/J.JACC.2017.01.060. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Pedersen SF, Sandholt BV, Keller SH, Hansen AE, Clemmensen AE, Sillesen H, Højgaard L, Ripa RS, Kjær A. 64Cu-DOTATATE PET/MRI for Detection of Activated Macrophages in Carotid Atherosclerotic Plaques: Studies in Patients Undergoing Endarterectomy. Arterioscler Thromb Vasc Biol. 2015;35:1696–1703. doi: 10.1161/ATVBAHA.114.305067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Malmberg C, Ripa RS, Johnbeck CB, Knigge U, Langer SW, Mortensen J, Oturai P, Loft A, Hag AM, Kjær A. 64Cu-DOTATATE for Noninvasive Assessment of Atherosclerosis in Large Arteries and Its Correlation with Risk Factors: Head-to-Head Comparison with 68Ga-DOTATOC in 60 Patients. J Nucl Med. 2015;56:1895–1900. doi: 10.2967/jnumed.115.161216. [DOI] [PubMed] [Google Scholar]
- 34.Lee R, Kim J, Paeng JC, Byun JW, Cheon GJ, Lee DS, Chung J-K, Kang KW. Measurement of 68Ga-DOTATOC Uptake in the Thoracic Aorta and Its Correlation with Cardiovascular Risk. Nucl Med Mol Imaging (2010). 2018;52:279–286. doi: 10.1007/s13139-018-0524-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Döring Y, Pawig L, Weber C, Noels H. The CXCL12/CXCR4 chemokine ligand/receptor axis in cardiovascular disease. Front Physiol. 2014;5:212. doi: 10.3389/fphys.2014.00212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hyafil F, Pelisek J, Laitinen I, et al. Imaging the Cytokine Receptor CXCR4 in Atherosclerotic Plaques with the Radiotracer 68Ga-Pentixafor for PET. J Nucl Med. 2017;58:499–506. doi: 10.2967/jnumed.116.179663. [DOI] [PubMed] [Google Scholar]
- 37.Weiberg D, Thackeray JT, Daum G, Sohns JM, Kropf S, Wester H-J, Ross TL, Bengel FM, Derlin T. Clinical Molecular Imaging of Chemokine Receptor CXCR4 Expression in Atherosclerotic Plaque Using 68Ga-Pentixafor PET: Correlation with Cardiovascular Risk Factors and Calcified Plaque Burden. J Nucl Med. 2018;59:266–272. doi: 10.2967/jnumed.117.196485. [DOI] [PubMed] [Google Scholar]
- 38.Li X, Heber D, Leike T, Beitzke D, Lu X, Zhang X, Wei Y, Mitterhauser M, Wadsak W, Kropf S, Wester HJ, Loewe C, Hacker M, Haug AR. [68Ga]Pentixafor-PET/MRI for the detection of Chemokine receptor 4 expression in atherosclerotic plaques. Eur J Nucl Med Mol Imaging. 2018;45:558–566. doi: 10.1007/s00259-017-3831-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Derlin T, Sedding DG, Dutzmann J, et al. Imaging of chemokine receptor CXCR4 expression in culprit and nonculprit coronary atherosclerotic plaque using motion-corrected [68Ga]pentixafor PET/CT. Eur J Nucl Med Mol Imaging. 2018;45:1934–1944. doi: 10.1007/s00259-018-4076-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Laitinen I, Marjamäki P, Någren K, Laine VJO, Wilson I, Leppänen P, Ylä-Herttuala S, Roivainen A, Knuuti J. Uptake of inflammatory cell marker [11C]PK11195 into mouse atherosclerotic plaques. Eur J Nucl Med Mol Imaging. 2009;36:73–80. doi: 10.1007/s00259-008-0919-6. [DOI] [PubMed] [Google Scholar]
- 41.Gaemperli O, Shalhoub J, Owen DRJ, Lamare F, Johansson S, Fouladi N, Davies AH, Rimoldi OE, Camici PG. Imaging intraplaque inflammation in carotid atherosclerosis with 11C-PK11195 positron emission tomography/computed tomography. Eur Heart J. 2012;33:1902–1910. doi: 10.1093/eurheartj/ehr367. [DOI] [PubMed] [Google Scholar]
- 42.Hellberg S, Silvola JMU, Kiugel M, et al. 18-kDa translocator protein ligand 18F-FEMPA: Biodistribution and uptake into atherosclerotic plaques in mice. J Nucl Cardiol. 2017;24:862–871. doi: 10.1007/s12350-016-0527-y. [DOI] [PubMed] [Google Scholar]
- 43.Hellberg S, Liljenbäck H, Eskola O, Morisson-Iveson V, Morrison M, Trigg W, Saukko P, Ylä-Herttuala S, Knuuti J, Saraste A, Roivainen A. Positron Emission Tomography Imaging of Macrophages in Atherosclerosis with 18 F-GE-180, a Radiotracer for Translocator Protein (TSPO). Contrast Media Mol Imaging. 2018;2018:1–11. doi: 10.1155/2018/9186902. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Kopecky C, Pandzic E, Parmar A, Szajer J, Lee V, Dupuy A, Arthur A, Fok S, Whan R, Ryder WJ, Rye K-A, Cochran BJ. Translocator protein localises to CD11b+ macrophages in atherosclerosis. Atherosclerosis. 2019;284:153–159. doi: 10.1016/J.ATHEROSCLEROSIS.2019.03.011. [DOI] [PubMed] [Google Scholar]
- 45.van Dongen GAMS Visser GWM, Lub-de Hooge MN, de Vries EG, Perk LR. Immuno-PET: a navigator in monoclonal antibody development and applications. Oncologist. 2007;12:1379–1389. doi: 10.1634/theoncologist.12-12-1379. [DOI] [PubMed] [Google Scholar]
- 46.Varasteh Z, Mohanta S, Li Y, López Armbruster N, Braeuer M, Nekolla SG, Habenicht A, Sager HB, Raes G, Weber W, Hernot S, Schwaiger M. Targeting mannose receptor expression on macrophages in atherosclerotic plaques of apolipoprotein E-knockout mice using 68Ga-NOTA-anti-MMR nanobody: non-invasive imaging of atherosclerotic plaques. EJNMMI Res. 2019;9:5. doi: 10.1186/s13550-019-0474-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.De Vos J, Mathijs I, Xavier C, Massa S, Wernery U, Bouwens L, Lahoutte T, Muyldermans S, Devoogdt N. Specific Targeting of Atherosclerotic Plaques in ApoE−/− Mice Using a New Camelid sdAb Binding the Vulnerable Plaque Marker LOX-1. Mol Imaging Biol. 2014:690–698. doi: 10.1007/s11307-014-0731-6. [DOI] [PubMed] [Google Scholar]
- 48.Broisat A, Hernot S, Toczek J,et al. Nanobodies Targeting Mouse/Human VCAM1 for the Nuclear Imaging of Atherosclerotic Lesions. Circ Res. 2012;110:927–937. doi: 10.1161/CIRCRESAHA.112.265140. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Bala G, Blykers A, Xavier C, et al. Targeting of vascular cell adhesion molecule-1 by 18F-labelled nanobodies for PET/CT imaging of inflamed atherosclerotic plaques. Eur Heart J Cardiovasc Imaging. 2016;17:1001–1008. doi: 10.1093/ehjci/jev346. [DOI] [PubMed] [Google Scholar]
- 50.Senders ML, Hernot S, Carlucci G, et al. Nanobody-Facilitated Multiparametric PET/MRI Phenotyping of Atherosclerosis. JACC Cardiovasc Imaging. 2019;12:2015–2026. doi: 10.1016/J.JCMG.2018.07.027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Que X, Hung M-Y, Yeang C, et al. Oxidized phospholipids are proinflammatory and proatherogenic in hypercholesterolaemic mice. Nature. 2018;558:301–306. doi: 10.1038/s41586-018-0198-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Senders ML, Que X, Cho YS, et al. PET/MR imaging of malondialdehyde-acetaldehyde epitopes with a human antibody detects clinically relevant atherothrombosis. J Am Coll Cardiol. 2018;71:321–335. doi: 10.1016/j.jacc.2017.11.036. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.De Groeve K, Deschacht N, De Koninck C, Caveliers V, Lahoutte T, Devoogdt N, Muyldermans S, De Baetselier P, Raes G. Nanobodies as tools for in vivo imaging of specific immune cell types. J Nucl Med. 2010;51:782–789. doi: 10.2967/jnumed.109.070078. [DOI] [PubMed] [Google Scholar]
- 54.Rashidian M, Ingram JR, Dougan M, Dongre A, Whang KA, LeGall C, Cragnolini JJ, Bierie B, Gostissa M, Gorman J, Grotenbreg GM, Bhan A, Weinberg RA, Ploegh HL. Predicting the response to CTLA-4 blockade by longitudinal noninvasive monitoring of CD8 T cells. J Exp Med. 2017;214:2243–2255. doi: 10.1084/jem.20161950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Krasniqi A, D’Huyvetter M, Devoogdt N, Frejd FY, Sörensen J, Orlova A, Keyaerts M, Tolmachev V. Same-Day Imaging Using Small Proteins: Clinical Experience and Translational Prospects in Oncology. J Nucl Med. 2018;59:885–891. doi: 10.2967/jnumed.117.199901. [DOI] [PubMed] [Google Scholar]
- 56.Mulder WJM, Van Leent MMT, Lameijer M, Fisher EA, Fayad ZA, Pérez-Medina C. High-Density Lipoprotein Nanobiologics for Precision Medicine. Acc Chem Res. 2018;51:127–137. doi: 10.1021/acs.accounts.7b00339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Pérez-Medina C, Binderup T, Lobatto ME, et al. In Vivo PET Imaging of HDL in Multiple Atherosclerosis Models. JACC Cardiovasc Imaging. 2016;9:950–961. doi: 10.1016/j.jcmg.2016.01.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Keliher EJ, Ye Y-X, Wojtkiewicz GR, et al. Polyglucose nanoparticles with renal elimination and macrophage avidity facilitate PET imaging in ischaemic heart disease. Nat Commun. 2017;8:14064. doi: 10.1038/ncomms14064. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Beldman TJ, Senders ML, Alaarg A, et al. Hyaluronan Nanoparticles Selectively Target Plaque-Associated Macrophages and Improve Plaque Stability in Atherosclerosis. ACS Nano. 2017;11: 5785–5799. doi: 10.1021/acsnano.7b01385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Binderup T, Duivenvoorden R, Fay F, et al. Imaging-assisted nanoimmunotherapy for atherosclerosis in multiple species. Sci Transl Med. 2019;11:eaaw7736. doi: 10.1126/scitranslmed.aaw7736. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Fayad ZA, Mani V, Woodward M, Kallend D, Abt M, Burgess T, Fuster V, Ballantyne CM, Stein EA, Tardif J-C, Rudd JHF, Farkouh ME, Tawakol A, dal-PLAQUE Investigators. Safety and efficacy of dalcetrapib on atherosclerotic disease using novel non-invasive multimodality imaging (dal-PLAQUE): a randomised clinical trial. Lancet. 2011;378:1547–1559. doi: 10.1016/S0140-6736(11)61383-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.
