Next Article in Journal
Molecular Basis Underlying the Therapeutic Potential of Vitamin D for the Treatment of Depression and Anxiety
Previous Article in Journal
Glucose Stimulates Glial Cell Line-Derived Neurotrophic Factor Gene Expression in Microglia through a GLUT5-Independent Mechanism
Previous Article in Special Issue
The Promises of Natural Killer Cell Therapy in Endometriosis
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis

by
Fatemeh Khodadust
1,
Aiarpi Ezdoglian
1,
Maarten M. Steinz
1,
Judy R. van Beijnum
2,
Gerben J. C. Zwezerijnen
3,
Gerrit Jansen
1,
Sander W. Tas
4 and
Conny J. van der Laken
1,*
1
Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
2
Angiogenesis Laboratory, Department of Medical Oncology, Amsterdam University Medical Center, Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
3
Department of Radiology and Nuclear Medicine, Amsterdam University Medical Center, Location VUmc, De Boelelaan 1117, 1081 HV Amsterdam, The Netherlands
4
Department of Rheumatology and Clinical Immunology, Amsterdam University Medical Center, Location AMC, Mijbergdreef 9, 1105 AZ Amsterdam, The Netherlands
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(13), 7071; https://doi.org/10.3390/ijms23137071
Submission received: 1 June 2022 / Revised: 23 June 2022 / Accepted: 23 June 2022 / Published: 25 June 2022
(This article belongs to the Special Issue Innovative Targeted Therapies in Inflammatory Diseases)

Abstract

:
Extensive angiogenesis is a characteristic feature in the synovial tissue of rheumatoid arthritis (RA) from a very early stage of the disease onward and constitutes a crucial event for the development of the proliferative synovium. This process is markedly intensified in patients with prolonged disease duration, high disease activity, disease severity, and significant inflammatory cell infiltration. Angiogenesis is therefore an interesting target for the development of new therapeutic approaches as well as disease monitoring strategies in RA. To this end, nuclear imaging modalities represent valuable non-invasive tools that can selectively target molecular markers of angiogenesis and accurately and quantitatively track molecular changes in multiple joints simultaneously. This systematic review summarizes the imaging markers used for single photon emission computed tomography (SPECT) and/or positron emission tomography (PET) approaches, targeting pathways and mediators involved in synovial neo-angiogenesis in RA.

1. Introduction

Rheumatoid arthritis (RA) is a chronic systemic autoimmune disease with an approximate prevalence of 1% of the adult population worldwide. This debilitating heterogeneous disease is characterized by the onset of synovial angiogenesis and inflammation, which eventually leads to the hyperplasia of the synovial lining and joint destruction [1,2,3]. Continuous vascular activity is found to promote cartilage and bone destruction, even when patients are in clinical remission [4].
The healthy synovium is characterized by a unique lining/sublining architecture. There is a delicate intimal lining composed of 1–2 layers of macrophage-like (type A) and fibroblast-like (type B) synoviocytes. Upon inflammatory triggering, the synovium undergoes a transitory pre-vascular stage in which the lining layer expands into sublining through fibroblast growth and extracellular matrix deposition (Figure 1) [5]. Simultaneously, the sublining is occupied by inflammatory cells (macrophages, T cells, and B cells). These events result in synovial tissue proliferation, called pannus, which acts like a local tumor, causing bone erosions observed in RA. In response to the increased requirement of oxygen and nutrients caused by the overpopulation of cells in the synovium, the vascular stage rapidly arises. This stage usually takes place at the time of RA clinical diagnosis. The increased density and permeability of blood vessels boosts synovitis by allowing immune cells to emigrate from the blood into the inflamed synovium, which in turn favors chronicity [3,6,7].
Angiogenesis is a complex multifaceted process that is defined as the formation of new blood vessels from the pre-existing vasculature via sprouting or remodeling. This is distinct from vasculogenesis and neovascularization, which refer to the de novo production of vascular networks from endothelial precursor cells [8,9]. Angiogenesis is involved in several physiological and pathological conditions including various inflammatory diseases such as RA, psoriasis, and atherosclerosis as well as cancer [10]. Under normal conditions, angiogenesis is a self-controlled and self-limiting process with balance in pro/anti-angiogenic factors [7,11,12,13]. In pathological angiogenesis, however, this balance is impaired and causes continuous endothelial cell (EC) activation, basement membrane rupture and migration, followed up by EC expansion, lumen formation and, finally, the formation of new blood vessels [14,15]. Here, the angiogenic process offers a variety of target structures for diagnostic tools and therapeutic interventions [6].
In RA, angiogenesis mainly relies on the dynamic, temporally, and spatially interaction of vascular ECs, fibroblasts, macrophages, and the extracellular matrix (Figure 1) [7,11,12]. Synovial fibroblasts and activated immune cells are responsible for the production of a large number of inflammatory cytokines, which are believed to play a crucial role in the development and progression of RA. Among the pro-inflammatory cytokines, TNF-α, IL-1, IL-6, and IL-8 are the principal cytokines that regulate the formation of other inflammatory mediators in the synovial tissue. These cytokines exert pro-angiogenic effects in RA both through their direct effect on ECs (meaning the response of ECs to the cytokine results in new vessel formation) as well as their indirect effect (by subsequent release of factors that induce endothelial cells angiogenesis) in the inflamed RA synovium producing pro-angiogenic factors including vascular EC growth factor (VEGF) [10,16,17]. VEGF and its receptors are one of the most extensively studied and fundamental players in the regulation of EC proliferation, angiogenesis, and capillary hyper-permeability. The pro-inflammatory cytokine networks stimulate synovial fibroblasts and other cells to release VEGF and other growth factors. Elevated levels of VEGF have been found in the plasma and synovial fluid of RA patients [18]. Moreover, correlations between VEGF serum level and inflammatory parameters of RA including erythrocyte sedimentation rate (ESR), C-reactive protein (CRP), and disease activity score 28 (DAS28) have been reported [19,20,21,22,23,24]. In the synovial angiogenesis cascade of events, other important mediators have also been recognized, which mostly involve matrix-degrading proteases, cellular adhesion receptors as well as basal membrane and interstitial matrix macromolecules (Figure 1) [16].
At present, there is no cure for RA and current treatment strategies mainly attempt to achieve remission or a low disease activity state (LDAS) as much as possible [1,2]. Three classes of therapeutic agents are commonly used in RA treatment: classical disease-modifying anti-rheumatic drugs (cDMARDs) including steroids, targeted synthetic (tDMARDs) and biological (bDMARDs), which can clinically mitigate the severity of RA, attenuate disease progression, and inhibit subsequent joint damage. None of these therapies specifically target angiogenesis in RA. However, several of these c/t/bDMARDs have been reported to partially impact angiogenesis at different levels [3,25,26,27,28,29,30,31,32,33]. For instance, blocking of the JAK/STAT-pathway by tofacitinib inhibited the formation of new synovial vessels [31]. Furthermore, some clinical studies and preclinical models of arthritis have revealed the benefits of anti-angiogenic therapies [31,33]. Notably, the proposed anti-angiogenic agents for RA were originally developed for cancer therapy and exploit some biological analogies between cancers and synovial pannus [7,34]. Due to the central position of angiogenesis in RA synovitis, therapeutic targeting of synovial angiogenesis by novel anti-angiogenic drugs may offer new opportunities for RA treatment.
The optimal management of RA relies on tools that can support clinical assessment in early and accurate disease detection, the prediction of prognosis, and monitoring of treatment response [35]. Diagnosing RA can be complicated due to the heterogeneous nature of the disease [1,36]. Although no detailed diagnostic criteria exist, the classification criteria of the American College of Rheumatology/European League Against Rheumatism [ACR/EULAR] Rheumatoid Arthritis Classification criteria 2010 [37] serve as a standard format to support clinical diagnosis. These criteria are based on clinical signs and serological assays (autoantibody and acute-phase reactant levels) and incorporate imaging techniques such as conventional radiography, ultrasound (US) and magnetic resonance imaging (MRI) for the detection of synovitis to improve earlier diagnosis and correct classification of patients [1,38,39].
Molecular imaging may offer additional opportunities for early RA disease activity assessment, prediction of treatment response, and monitoring during treatment. Over the past decade, several noninvasive molecular imaging techniques such as conventional planar (2D) scintigraphy and 3-dimensional (3D) single photon emission computed tomography (SPECT), positron emission tomography (PET), and hybrid modalities have been developed and applied for RA diagnostic purposes [40,41]. These modalities rely on the in vivo biodistribution and quantification of the radiotracer binding to a specific molecular target in the body, allowing for exceptional target specificity and sensitivity [42,43]. Traditional 2D scintigraphy and SPECT are imaging techniques that use gamma cameras to detect gamma rays emitted by radionuclides. Unlike planar scintigraphy, SPECT provides 3D tomographic images and yield slices through the body. Typical radionuclides used for planar scintigraphy and SPECT imaging are technetium 99mTc (t1/2: 6.0 h), indium 111In (t1/2: 2.8 days), and iodine 123I (t1/2: 13.2 h) [44,45]. PET adopted different isotopes (positrons) for imaging and holds certain superiority, resulting in increased popularity both in preclinical and clinical settings. It provides improved imaging quality with higher diagnostic accuracy and sensitivity, allowance for reliable quantification, and mostly a lower radiation burden to patients. Most used positron-emitting radioisotopes clinically and/or pre-clinically include fluorine 18F (t1/2: 110 min), carbon 11C (t1/2: 20 min), copper 61/64Cu (t1/2: 3.3 h and 12.7 h), gallium 66/68Ga (t1/2: 9.5 and 1.1 h), zirconium 89Zr (t1/2: 78.4 h), and iodine 124I (t1/2: 100.2 h) [45,46]. These radionuclides can be conjugated with an appropriate ligand with available binding sites. Depending on the type of radionuclide (e.g., metallic or non-metallic) and/or achieving the desirable biological activity of a targeting vector, the chelator and/or linker (spacer) can be utilized with which the in vivo biological behavior of the radioligand can be significantly affected [47]. The structural components of radiopharmaceuticals discussed below in the context of angiogenesis research and applications commonly comply with the four compartment criteria depicted in Figure 2A [44,48]. Targeting vector can be bound to a radionuclide and ensures accumulation into the target cells. Linkers and chelators can be used to attach the radionuclide and targeting vector. Some radionuclides, particularly radiometals (such as 99mTc, 66/68Ga), require chelators to form stable complexes. Linkers or spacers are often optional components and usually are used to improve the pharmacokinetics of a radiotracer and/or reduce the steric interference and retain high binding affinity [44,47,48]. For clinical utility, it is of importance that a radiotracer meets some basic requirements. In this regard, ideal radiotracers should harbor the properties indicated in Figure 2B [49].
Although both SPECT and PET techniques have deep signal penetration and demonstrate high sensitivity in molecular targets/processes imaging, their spatial resolution is not as high as CT or MRI. However, high contrast resolution and the capability of providing functional images make these two imaging modalities highly attractive, and new technological improvements in PET scanners (total body PET) allow for significant improvement in spatial resolution [50,51]. The feasibility of hybrid imaging systems has facilitated the combination of high sensitivity radionuclide imaging with high-resolution CT imaging, allowing for co-localization of molecular images with anatomical images. PET/MR has important (pre)clinical potential by also providing soft tissue morphological imaging. Ultimately, these imaging approaches may contribute to better-individualized health care as physicians are informed by more accurate and quantified molecular signals within specific anatomical structures of interest [42]. The use of molecular imaging tools may facilitate specific drug development and provide guidance in personalized medicine [1,52,53].
It is worth mentioning that molecular imaging modalities, beyond nuclear imaging techniques, also consist of two other non-ionizing main categories: optical and acoustic (ultrasound) imaging. Although optical techniques such as bioluminescence and fluorescence imaging as well as acoustic imaging provide a highly versatile platform for noninvasive in vivo molecular imaging [54,55], they were beyond the scope of this paper. This systematic review is focused on non-invasive in vivo nuclear imaging.
Concerning angiogenesis imaging, computed tomography (CT) and magnetic resonance imaging (MRI) may not be suitable to assess the response to anti-angiogenic treatment since these techniques merely allow for the evaluation of morphological and perfusion parameters, but do not enable the depiction of changes in the vasculature at the molecular level [15,53]. 18F-FDG PET/CT is a sensitive technique for evaluating disease activity and treatment response in patients with RA [56]. However, the mechanism underlying 18F-FDG uptake is rather unspecific and is largely associated with elevated glucose metabolism in inflamed cells and tissues [57]. In the case of visualizing angiogenesis in early, developing RA for diagnostic purposes and monitoring anti-angiogenetic properties of RA drugs, molecular imaging markers that target synovial angiogenesis are needed.
The objective of this systematic review was to present an overview of all the potential molecular imaging tracers (PET and SPECT/scintigraphy) involved in the RA angiogenic cascade that have been studied in pre-clinical and/or clinical settings in RA.

2. Methods

2.1. Eligibility Criteria

This systematic review focused on studies investigating the detection of the pathological angiogenesis in RA through molecular imaging techniques. All studies on the application of radiotracers using PET and SPECT/scintigraphy for diagnosis, prognosis, and response assessment to therapy in preclinical and/or clinical stages of RA (related to angiogenesis) were included. No restrictions were placed on the type of studies included as research on exploiting angiogenesis for RA molecular imaging is still in development.

2.2. Information Source and Search Strategy

For this systematic review, a literature search was carried out using the following databases without date and language restriction until March 2022: PubMed, EMBASE, and the Cochrane Library. The literature was reviewed following the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines [58].
Two separate searches were conducted in consultation with the medical information specialists at the University Library VU (Vrije Universiteit Amsterdam); the initial search focused on all radiopharmaceuticals used for imaging in RA by applying the controlled vocabulary search terms and free-text terms (medical subject headings (MeSH) and relevant keywords) based on the following words: PET/SPECT/Scintigraphy imaging and arthritis. An additional search was performed to identify important angiogenesis biomarkers in RA, through which radiopharmaceuticals that may be useful for angiogenesis targeting in RA were selected. The full search strategies for all databases can be found in the Supplementary Materials as File S1 (RA angiogenesis biomarkers search) and File S2 (RA radiotracers search).

2.3. Study Selection

Both co-reviewers (F.K. and M.K.) independently screened all potentially relevant titles and abstracts for eligibility. The initial evaluation was based on title, keywords, and abstracts of the retrieved studies. If necessary, the full-text article was checked for the eligibility criteria. This evaluation was performed at Rayyan QCRI (Qatar Computing Research Institute) on a platform expediting the initial screening of abstracts and titles; it allows for the transfer of the citations obtained from different databases and provides an individual blinded assessment of articles with the ability to invite an unlimited number of collaborators [59]. Disagreements after an initial blinded evaluation were resolved through discussion. Following consensus, full-text versions of the studies that met the eligibility criteria and those that were not considered eligible were acquired and assessed for the final determination of inclusion.

2.4. Data Extraction and Assessment

Data from the studies included were extracted by two authors (F.K. and M.K.). The reviewers independently extracted data for each search (angiogenesis biomarkers and RA radiotracers) and then a third reviewer (A.E.) randomly evaluated the collected data. The initial data extraction for the angiogenesis biomarkers of RA included the purpose of study (exploratory, treatment, diagnosis), diagnostic or therapeutic interventions used, molecular targets, and clinical stage. For radiotracers in RA, we characterized articles according to the type (PET and SPECT/scintigraphy), name of the tracer, and molecular targets.

2.5. Search Results

The first literature search that focused on nuclear imaging in RA resulted in 356 studies. The selected articles included case reports, letters, and the investigation of the pharmacokinetics of the tracers as well as tracers in both animal and human trials. In the second search, which focused on angiogenesis targets in RA, 323 potentially related studies were included. This search included studies that explored the role of a biomarker(s)/pathway(s) in the angiogenesis process, or reported the mechanism of action (or target) of a therapeutic or diagnostic approach involved in the angiogenesis in RA in pre-clinical/clinical settings. Next, the final related articles were selected by matching the angiogenesis targets in both searches. The flow diagram of the search and selection process is presented in Figure 3A,B. The final total number of included articles in the results was 39 studies. The radiotracers and accompanying articles were divided based on the angiogenesis targets (Table 1).

3. Results and Discussion

  • Results
  • Radiotracers targeting angiogenesis in RA
The PET and SPECT/scintigraphy tracers included in this review targeted different levels of pathological angiogenesis in RA. The radiotracers were categorized based on the type of targets reported to be involved in RA angiogenesis (Figure 1). An overview of the radiotracers is presented in Table 1 and is discussed in detail below.

3.1. Environmental Factor (Hypoxia)

Enhanced angiogenic activity in the synovial vasculature of arthritic joints is associated with the onset of a hypoxic environment [10]. Many studies have revealed the connection between VEGF and hypoxia [99,100,101,102]. Excessive leukocyte migration into the RA joint and hyperplasia of the synovial lining cells confers joint microenvironment hypoxia, leading to the activation of the key transcription factor HIF-α (two isoforms, HIF-1α, HIF-2α). This transcription factor induces the expression and secretion of VEGF by macrophages and RA synovial tissue fibroblasts. During hypoxia, a positive feedback between the HIF-1α and VEGF pathways occurs, which regulates continuous angiogenesis. Hypoxia, in turn, can facilitate the expression of many pro-angiogenic and pro-inflammatory genes including the chemokines IL-8 (also known as CXC-chemokine ligand 8 or CXCL8), CC-chemokine ligand 20 (CCL20), and CXCL12 [10,103,104].
So far, radiolabeled azomycins (2-nitroimidazoles) such as 18F-FMISO (18F-Fluoromisonidazole) [60,61], 18F-FAZA (18F-fluoroazomycinarabinoside) [61], 123I-IAZA (1-(α-D-5-Iodoarabinofuranosyl)-2-nitroimidazole) [64], and more recently 64/67Cu-ATSM (copper bis-thiosemicarbazone complexes) [62] have been explored for hypoxia imaging in RA.
The HIF-1α tracers, 18F-FMISO and 18F-FAZA, are among the commonly clinically used tracers that have been investigated for the identification of tumor hypoxia in malignancies. These nitroimidazole compounds are metabolically trapped in tissues with low oxygen tensions [105]. PET studies using these hypoxia tracers in a glucose-6-phosphate isomerase (GPI)-induced arthritis mouse model revealed an increased uptake in the arthritic joints with comparable arthritis-to-control joint ratios (ratio 18F-FMISO: 2.7; ratio 18F-FAZA: 2.8). However, 18F-FAZA exhibited a greater hydrophilicity and faster tissue clearance, and thus reduced whole-body background compared to 18F-FMISO. Histological studies, simultaneous MRI imaging, and clinical swelling score of the inflamed joints correlated well (especially for 18F-FMISO) with the uptake of the radiotracers. The utility of these PET tracers has been suggested for the detection of early pathological changes in the initial phases of RA before the onset of cartilage and joint destruction [61].
In another study, 18F-FMISO was used to explore the therapeutic potential of two different classes of novel MAPK inhibitors (the highly selective p38 MAPK inhibitor (Skepinone-L) and the dual p38 MAPK/JNK 3 inhibitor (LN 950) in the K/BxN serum transfer model of RA [60]. MAPK activity is associated with hypoxia and is a crucial factor in RA progression through the propagation of angiogenesis and pannus formation. Both inhibitors markedly suppressed arthritic disease activity, but the radiotracer uptake in the ankles of skepinone-L-treated mice did not reflect the clinical parameter of ankle swelling very well. It was concluded that this hypoxia targeting tracer might not be a reliable tool to monitor MAPK inhibition when compared to the parameter of ankle swelling, although the MAPK inhibitors directly influence hypoxia via the modulation of cell proliferation and angiogenesis [60].
The in vivo behavior of another hypoxia radiotracer, 64/67Cu-ATSM, which is mainly used in oncology, cardiology, and neurology, has recently been assessed in the collagen-induced arthritis (CIA) mouse model [62]. The 64/67Cu-ATSM study data exhibited a correlation between the accumulation level of the tracer and the clinical scores of the arthritic paws. However, when validating the specificity of the targeting hypoxia, hypoxic tissue conditions of the inflamed joints were not confirmed by the analysis of other markers for hypoxia [62].
123I-IAZA has also been developed for the detection of tissue hypoxia [63]. Nevertheless, in a clinical study with scintigraphic evidence of arthritic joint hypoxia, the uptake intensity of this radiotracer did not correlate with the disease severity in RA patients [64].

3.2. Somatostatin Receptors—Somatostatin

Somatostatin is a natural, widely distributed, small cyclic peptide that inhibits exocrine secretion, cellular proliferation, and cellular differentiation and promotes apoptosis. The effects of somatostatin and its analogues are mediated via five (G-protein-linked) receptors (SST 1, 2, 3, 4, 5). These receptors regulate multiple signal transduction pathways to elicit their inhibitory effects [106]. The modulatory effects on immune response, anti-proliferative, anti-angiogenic, and analgesic properties of somatostatin and its analogues have been explored [107]. Both the activated lymphocytes and inflamed vascular endothelium can express SSTR 2, 3, and 5 in different physiological and pathologic conditions [108].
Some studies have shown that SST may have a functional role in angiogenesis. It has been reported that SST2 is particularly expressed in the angiogenic sprouts of endothelial cells (HUVEC) in vitro, and experimental angiogenesis is inhibited by the synthetic somatostatin analogue (e.g., octreotide) [106,109,110,111]. In RA, somatostatin agonists have been demonstrated to potently abrogate VEGF levels as well as pro-angiogenic activity. Additionally, SST2 is uniquely upregulated during the angiogenic cascade. The receptor exists in two isoforms: SST2A and SST2B. The isotype SST2A is expressed at a high density during inflammatory response in arthritis mouse models. Synovial biopsies from RA patients have demonstrated the presence of SST2A in the synovial venule endothelium as well as on a subset of synovial macrophages [112]. In vitro, somatostatin treatment on RA synoviocyte cells was associated with the downregulation of MMP. Specifically MMP-1, MMP-2, and MMP-9 are known to trigger the angiogenic cascade [113]. These characteristics make somatostatin/SSTs attractive candidates in the context of angiogenesis for both diagnostic and therapeutic use in immune-mediated diseases such as RA [108].
In an original study, Vanhagen et al. reported the use of somatostatin scintigraphy for the visualization of clinically affected joints in RA patients [67]. The results demonstrated that the SST2-preferring analogue 111In-pentetreotide (or [111In-DTPA-D-Phe1]-octreotide) accumulated in active rheumatoid joints, but not in osteoarthritic joints [67].
In another clinical study in 18 RA patients, a radiolabeled somatostatin analogue (99mTc-EDDA/HYNIC-TOC) was used to evaluate the activity status of joint inflammation. The patients were also scanned after receiving infliximab (anti-TNF-α) therapy [66]. The results showed intense uptake with a symmetric and focal pattern in the hands, predominantly in the carpal, metacarpal, and proximal interphalangeal joints. All patients and all positive joints showed a clinical and scintigraphic improvement after infliximab therapy. No correlation was observed between joint pain or swelling and somatostatin receptor scintigraphy positivity. Some joints that were apparently clinically poorly positive showed high somatostatin uptake in the images (potentially pointing at subclinical disease detection) and vice versa. The targeting was explained to be associated with the overexpression of somatostatin receptors in active phases of the disease, which is characterized by endothelial activation and lymphocyte infiltration in the synovium [66,114].
The 99mTc-depreotide is a 99mTc-labeled somatostatin analogue with a high affinity for subtypes 2, 3, and 5 of somatostatin receptors [68]. In a study, this tracer exhibited good sensitivity to localize in the foci of active osteomyelitis, however, it was not specific and also could detect recent osteonecrosis and RA. 99mTc-depreotide scintigraphy was proposed as a complementary method for the investigation of bone infection and inflammation, particularly osteomyelitis and cases of recent osteonecrosis and RA. The authors postulated that since these particular receptors are overexpressed on the surface of activated leucocytes, the detection of the inflammation by this tracer can be associated with the increased vascular supply and permeability, along with considerable leucocyte infiltration [68].
Shamim et al. conducted a clinical study for a comparative evaluation of the conventional MDP bone scan and 68Ga-labeled SST analogue DOTA-NOC PET/CT in the articular manifestation of RA [65]. The radiolabeled SST analogue did not exhibit superiority over the MDP scan scintigraphy for the detection of the clinically diagnosed joints for RA. SSTR PET/CT and MDP scans were almost equally able to detect the RA affected joints with a detection rate of 77% and 80% of clinically positive joints, respectively. Nevertheless, this radiotracer can be considered as a theranostic tool for RA patients [65].

3.3. Matrix Metalloproteases

Matrix metalloproteinases (MMPs) are a subfamily of zinc-dependent enzymes and on the basis of their substrates and the organization of their structural domains are classified into collagenases, gelatinases, stromelysins, matrilysins, membrane-type (MT)-MMPs, and other MMPs [115]. They can be secreted by various cell types such as fibroblasts, vascular smooth muscle, and leukocytes and contribute to tissue remodeling during various physiological and pathological processes [115,116]. MMPs promote EC migration and tube formation by proteolytically remodeling the basement membrane [117]. Their distribution is restricted in normal joint tissue; however, in response to inflammatory cytokines and the production of growth factors, MMPs are expressed in the synovium and cartilage. In RA, MMP-3 is mainly produced by synovial lining layer cells and by ECs, while MMP-9 (gelatinase B) and MMP-13 (collagenase 3) proteins are expressed mainly by ECs, fibroblasts, chondrocytes, and leukocytes within the inflamed synovium. Membrane type 1-MMP (MT1-MMP) mRNA has been detected in ECs and fibroblasts [118,119]. MMPs contribute to angiogenesis by breaking down the basement membrane and other ECM components, not only allowing ECs to detach and migrate into new tissue, but also by releasing ECM-bound proangiogenic factors (bFGF, VEGF, and TGFβ) [120,121,122].

MMP-2/-9/-13

Among the MMPs, MMP-9 and MMP-13 have received specific focus for their proactive role in the angiogenic process in the arthritic joint. Research on the association of MMP-2 and MMP-9 and MMP-1 and MMP-13 with VEGF in the synovial fluid of RA patients has shown that only the MMP-9 and MMP-13 levels were significantly correlated with the VEGF levels [10,123]. Moreover, a strong positive association was observed between MMP-13 with MMP-9 and urokinase plasminogen activator (uPA). Among the various MMPs, MMP-9 and MMP-13 cooperate in joint degradation, and the expression levels of both factors were markedly elevated in the sera and joints of RA patients when compared to osteoarthritis (OA) and associated with disease progression and severity. The potential role of uPA in angiogenesis through the activation of MMP-13 has also been outlined [124]. Based on the association between MMP-9 and VEGF in RA patients [123] and the reduced severity of antibody-induced arthritis in MMP-9 knockout mice [125], it is assumed that the inhibition of MMP-9 can attenuate angiogenesis in the pannus and inhibit the activation of proMMP-13 (inactive MMP-13) in the arthritic joint [124]. The membrane-type matrix metalloproteinases (MT-MMPs) have shown to cleave and activate other MMPs such as MMP-2 and MMP-13. In MT1-MMP (MMP-14) knockout mice, the severe phenotype with impaired angiogenesis has been suggested to occur due to the lack of the activation of MMP-13 and MMP-9 [126].
Breyholz et al. evaluated a series of barbiturate derivatives (pyrimidine-2,4,6-triones (RO 28-2653)) as a new class of potential selective MMP-inhibitor (MMPI) radiotracers by radiolabeling them with iodine-125 [127]. Indeed, RO 28-2653 elicited antiangiogenic efficacy due to the inhibitory activities on MMP-2, MMP-9, and MMP-14. These observations supported the approach of using barbiturates as a radiotracer to visualize angiogenesis [127,128]. Structural modifications have been performed in the barbiturate derivative by converting it to a more hydrophilic form using a mini polyethylene glycol (PEG) linker. Preliminary biodistribution studies with one of these modified structures (18F-26) indicated no tissue-specific accumulation in wild-type mice [128]. In an extension to this work, a new series of fluorinated pyrimidine 2,4,6-trione-based MMPIs derivates (e.g., 18F-30) was identified as potent inhibitors of MMP-2, -8, -9, and -13. The radiotracer 18F-30 exhibited excellent stability in vitro and rapid clearance, as shown by in vivo biodistribution studies in wild-type mice [69]. The approach of barbiturate-based MMP-targeted radiotracers was proposed to be a viable non-invasive approach of in vivo PET imaging of MMP-2 and MMP-9-associated diseases including arthritis.
More recently, N,N′-bis(4-fluoro-3-methylbenzyl)pyrimidine-4,6-dicarboxamide-based selective and potent MMP-13 inhibitors have been successfully synthesized and radiolabeled with 11C, 18F, and 68Ga [70]. Among them, the 11C-methylated compound 2d represented the most lipophilic radiotracer with predominant hepatobiliary excretion and remaining tracer accumulation over time in the liver. In contrast, gallium-labeled derivatives revealed improved clearance characteristics achieved by introducing a more hydrophilic DOTA moiety to the conjugate. Therefore, preclinical evaluation of these PET radiotracers in murine disease models expressing upregulated levels of MMP-13 (e.g., breast cancer and arthritis) has been proposed [70].

3.4. Extracellular Matrix Proteins

Extracellular matrix (ECM) components including matrix molecules within the EC basement membrane such as type I collagen, laminin, fibronectin, vitronectin, tenascin, and various proteoglycans have been implicated in EC migration during angiogenesis [16]. The ECM controls EC activities by diverse mechanisms ranging from cell anchorage, integrin-mediated activation, and signaling to the binding, release, and activation of soluble growth factors and alteration in the supramolecular matrix architecture [129].

Extra Domain A (ED-A)

Fibronectin (FN) is an ECM protein that binds to integrins and to other ECM proteins. Fibronectin polymorphic variations result in the expression of three different domains [130]. Extra domain-A and -B (ED-A/-B) isoforms are recognized as important angiogenic markers that are synthesized, secreted, and deposited to the ECM structures from different cell types residing in the synovial lining layer. Studies have been conducted to determine the functional roles of the ED-A and ED-B domains of FN. The ED-A domain is involved in various processes including cell adhesion, myofibroblast differentiation, wound healing, cell cycle progression, and mitogenic signal transduction [131,132,133]. High expression levels of both isoforms have been detected in human pannus tissues on neovascular sites [132,134,135,136,137]. ED-A/-B have a restricted expression pattern in normal tissues, which is in contrast to fetal and transformed human tissues. Together, these findings have established fibronectin splicing variants as attractive targets for molecular imaging-based detection and monitoring/predicting the outcome of anti-angiogenic therapies [46].
Antibody-based pharmaco-delivery strategies, mainly studied in oncological settings, have also recently been proposed for use in RA. In this regard, the targeting properties and biodistribution of an ED-A binding antibody fragment (F8) conjugated with IL-10 (anti-inflammatory cytokine) were explored in preclinical and clinical settings via PET-guided imaging [138]. Iodine-124 labeled F8−IL10 accumulated not only in the clinically inflamed joints of patients, but also in the subclinically affected joints. These results were corroborated in an arthritis model in rats. Furthermore, this PET tracer showed rapid blood clearance and increased radioactivity in the spleen and liver. The radiotracers’ pharmacokinetic comparison in healthy and arthritic rats revealed higher uptake levels in the liver and spleen of the arthritic model. This indicates that the high uptake in these organs may be ED-A specific and seems to be associated with systemic inflammation. The encouraging results from F8-IL10 therapy studies [138] may hold promise for using this PET tracer to select patients who may benefit from this therapeutic approach [72].

3.5. Adhesion Molecules

Adhesion molecules are generally classified into five groups based on their molecular structure and mode of interaction [139] and members of each group have been detected in angiogenic blood vessels [140]. These groups include: (1) integrins (e.g., αVβ3, αVβ5, α5β1); (2) selectins (E/P-selectins); (3) proteins of the immunoglobulin superfamily (IgSF) including nectins and others such as mucins (e.g., VCAM-1, ICAM-1, JAM-A/C, Lewisy/H and MUC18); (4) cadherins [16,139,141]; and (5) proteins with enzymatic properties such as vascular adhesion protein 1 (VAP-1) [139,142] also play significant roles in perpetuating the inflammatory response and promoting angiogenesis. In fact, angiogenesis or vascular remodeling depends on both the growth factor stimulation and cell adhesion events [143]. The expression and function of vascular cell adhesion molecules (CAMs) are dynamically regulated following tissue alterations induced by the surrounding stromal changes. This property enables ECs to leave the quiescent state and re-enter the angiogenic cascade [144].

3.5.1. VAP-1

Vascular adhesion protein-1 (VAP-1/SSAO) is mainly expressed on vascular ECs, on smooth muscle cells, and on adipocytes. It is practically absent from the endothelial surface of normal tissues, while upon inflammation, it is rapidly translocated from the intracellular storage granules to the EC surface. VAP-1 contributes to several steps in the extravasation cascade and controls the trafficking of lymphocytes, granulocytes, and monocytes to sites of inflammation. Aside from being an adhesion molecule, it also harbors enzymatic properties [145,146]. MMPs can cleave VAP-1 and generate a soluble form of VAP-1 (designated sVAP-1 or semicarbazide sensitive amine oxidase, SSAO). Through its enzymatic action, VAP-1 oxidizes primary amines and produces hydrogen peroxide, aldehyde and ammonium, which can function as potent inflammatory mediators, leading to the upregulation of other adhesion molecules such as E- and P-selectin, ICAM-1, and VCAM-1. This enzymatic activity is indispensable for leukocyte extravasation through the endothelium. These unique features distinguish VAP-1 from other conventional adhesion molecules. Attempts have been made to elucidate the role of VAP-1 in the pathogenesis of arthritis in vivo, and for this purpose, several SSAO inhibitors of different classes have been developed and applied as potential therapeutic agents for both experimental and clinical use in arthritis [147,148,149,150,151,152]. However, it is still unclear whether the proangiogenic functions of VAP-1 merely rely on its ability to recruit VEGF-producing myeloid cells in different settings, or whether endothelial SSAO activity directly promotes neo-angiogenesis [146,151]. Interestingly, Siglec-9 and -10, both sialic acid-binding immunoglobulin-like lectins, have been identified as counter receptors for VAP-1 on B cells and granulocytes/monocytes, respectively [145,153,154].
Siglec-9 is expressed on all peripheral blood granulocytes where it can bind to the enzymatic groove of VAP-1. Siglec-9 expression is rapidly upregulated on the leukocyte surface after inflammatory stimuli such as TNF-α and lipopolysaccharide (LPS). TNF-α exposure increased Siglec-9-mediated granulocyte binding to the vasculature in the inflamed human synovium [145]. In order to analyze whether a Siglec-9 peptide could be used for in vivo inflammation imaging, it was radiolabeled with 68-gallium and its efficacy in the specific targeting of VAP-1 activated vasculature could be confirmed [145]. Gallium-68-labeled Siglec-9 has been explored for the assessment of synovitis in a rabbit model of synovitis (phytohemagglutinin-induced inflammation model) and the result was compared with 18F-FDG [74]. With 68Ga-DOTA-Siglec-9, the mild synovitis observed in this model was detected comparably to 18F-FDG (inflamed-to-control joint ratios were 1.2  ±  0.14 and 1.5  ±  0.4, respectively). Of note, VAP-1-positive vessels were found in the synovium, both in the PET images and immunohistochemistry (IHC) of the animal joints, which was considered to be due to the systemic response to the chemically-induced inflammation. Furthermore, it seems that there is a distinct VAP-1 expression pattern between humans and preclinical models of arthritis. In the arthritic animals, the mild synovitis induction and the level of vascular VAP-1 expression was lower when compared to that of the RA patients. In humans, VAP-1 expression on the vessel surface was not found in healthy synovial tissue [74].
The aforementioned observations in the rabbit synovitis model were consistent with the results of SPECT imaging using 123I-labeled fully human anti-VAP-1 monoclonal antibody BTT-1023 [106]. BTT-1023 also binds to the VAP-1 of other primates and rabbits but does not cross-react with rats or mice [75]. In this study, the distribution and pharmacokinetics were also assessed by PET/CT (through radiolabeling of the mAb with 124I) in healthy rabbits up to 72 h after injection. 124I-radioactivity accumulated in the thyroid gland and the liver. The tracer’s accumulation in these organs was attributed to the deiodination of the radiotracer and the previously mentioned different VAP-1 expression pattern in the animal models, respectively. All of the other organs had a low tracer uptake. The study revealed an inflammation-to-control joint uptake ratio of 1.2 ± 0.1 in the rabbits with arthritis. It was also mentioned that the estimated mean effective dose for a 70-kg man would be rather high for the PET version of this tracer as the estimated injected radioactivity of iodine-124 ideally needs to be higher than that of other PET radionuclides (as only 25% of the 124I decay is by positron emission). However, the estimated human radiation burden can be controlled by applying an adequate shielding and dose regimen [75].
Recently, the first-in-human study of 68Ga-DOTA-Siglec-9 was performed in order to detect early stage arthritis [73]. 68Ga-DOTA-Siglec-9 PET/CT and 18F-FDG PET/CT were performed in one patient with RA. The in vivo stability test of the tracers in plasma by HPLC analysis revealed, within 10 min, several peaks representing metabolites other than the radiotracer. The tracer pharmacokinetics performed in healthy men exhibited rapid renal clearance due to the hydrophilic and small size of the peptide [73]. A 30 min dynamic PET/CT scan of the hands of the RA patient showed uptake in the arthritic phalangeal joints. However, the observed SUVs of 68Ga-DOTA-Siglec-9 (0.7–1.05) were slightly lower than those of 18F-FDG (1.08–1.46). Although this radiotracer harbors promising properties, further optimization is required prior to application in large-scale clinical studies [73].

3.5.2. E-Selectin

Selectins are transmembrane proteins expressed on blood platelets and Weibel–Palade bodies of ECs (P-selectins), leukocytes (L-selectins), and ECs (E- and P-selectins). Selectins are involved in the recruitment of leukocytes from the bloodstream in a cascade-like manner into the inflamed tissue. E-selectin is not expressed under basal conditions [155]. Antibodies to soluble E-selectin block the chemotactic activity of rheumatoid synovial fluid on the ECs and its angiogenic activity both in vitro and in vivo [156]. Soluble E-selectin–induced angiogenesis is predominantly mediated through the SRC-phosphoinositide 3-kinases (PI3K) pathway [155], an important signaling pathway in the angiogenesis process [157]. Of note, a high expression of the inhibitor of DNA binding/differentiation (Id) in ECs within the RA synovial tissues was observed and in functional studies, the overexpression of Id promoted the expression of E-selectin [158]. The synovial fluid and serum levels of soluble E-selectin—together with bFGF—were significantly elevated in RA compared to the OA patients and the controls [159].
The murine 1.2B6 monoclonal antibody (mAb) has been found to selectively target E-selectin, and a Fab fragment of 1.2B6 labeled with 99mTc was compared with 111In-labeled 1.2B6 F(ab′)2 in RA patients using conventional bone scanning [77]. 111In-labeled 1.2B6 was previously validated in experimental arthritis in pigs and subsequently tested in clinical studies in RA. In preliminary studies in RA patients, the labeled 1.2B6 tracer showed higher specificity and better image contrast compared to HIG (nonspecific immunoglobulin tracer) coupled with either 111In or with 99mTc. However, the 111In-variant was not considered the tracer of choice due to practical reasons such as a higher radiation burden compared to Tc99m for further investigations [77,160]. Subsequently, 99mTc-labeled 1.2B6 Fab (99mTc-Fab) was demonstrated to be as effective as 111In-F(ab′)2. In fact, 99mTc-Fab can be used in a one-day protocol as the scan outcome appeared to be equivalent at 4 h compared with the 111In-F(ab′)2 scan outcome at 24 h in patients with RA. In addition, 99mTc Fab demonstrated more specific targeting of active joint inflammation compared with 99mTc-HDP [77].
Finally, nano-SPECT/CT imaging using 111In-1.2B6 was performed in a chimeric SCID mouse model as a novel tool for the pre-clinical development of radiopharmaceutical and delivery agents targeting human synovial tissue in vivo [76]. In this study, the uptake of this radiotracer was clearly visible in the human synovial tissue xenograft in the SCID mice at 24 h [76].

3.5.3. Integrins

Integrins are heterodimeric adhesion glycoproteins and comprise two different subunits, an α subunit and a β subunit. These subunits have particular ECM protein binding sites to regulate essential cellular survival, motility, migration, inflamed responses and invasion [161]. αvβ3 and α5β1, Arg-Gly-Asp (RGD) binding integrins, have been described in RA synovial tissue for their involvement in angiogenesis by promoting EC migration and survival, and their expression in fibroblasts, ECs, and synovial infiltrated cells [161]. The RGD sequence is a common cell recognition motif within the sequence of ECM proteins (e.g., vitronectin, fibronectin, fibrinogen etc.) and is required for interaction with integrins [162]. αvβ3 and α5β1 are recognized as fibronectin receptors. αvβ3 can also bind to vitronectin, fibronectin, osteopontin, and bone sialoprotein [162,163,164].
αvβ3 and α5β1 integrins can exert their angiogenic effects by regulating MMP expression. Growth factors, cytokines, hormones, and the composition of the ECM are able to activate integrins and alter their binding affinity. Once activated, several signaling cascades such as focal adhesion kinase (FAK), the mitogen-activated protein kinase (MAPK) pathway, the PI3K, and c-Jun N-terminal kinases (JNKs) are subsequently initiated, which in turn can induce the proliferation and migration of angiogenic ECs. Integrin αvβ3 is also known to work synergistically with VEGF to activate angiogenesis in ECs via VEGFR-2 phosphorylation [162,163,164]. Etaracizumab (vitaxin), a humanized mAb against αvβ3 integrin receptor, is the only antagonist that has entered phase II clinical trials for RA treatment. Despite showing good clinical benefits, further investigations were halted, probably due to the side effects observed and the limited efficacy of anti-angiogenic factors alone in controlling the disease progression [162,163,165].
Considering the potential of integrins as therapeutic targets for the treatment of RA, pinpointing the role of integrins in angiogenesis has been subject of a multitude of studies. PEG-HM-3, a novel integrin inhibitor, displayed effective anti-rheumatic activity in antigen-induced arthritis (AIA) and CIA animal models either as single agents or in combination therapy with methotrexate [166]. Another study demonstrated that urokinase-type plasminogen activator receptor (uPAR) could alter the biological characteristics of RA fibroblast-like synoviocytes via the β1-integrin/PI3K/Akt signaling pathway and thereby affect the neo-angiogenesis of synovial tissues in RA patients [167]. αvβ3, β1-integrins predominantly localize to vascular endothelium and lining layer cells in RA synovial tissue, in contrast to OA and normal control synovial tissue. Of note, acute-phase serum amyloid A (A-SAA) was found to increase αvβ3 and β1 binding in RA synovial fibroblasts [168]. The role of IL-18 as an angiogenic mediator in RA was reported to act via αvβ3 integrin and independently on the contribution of local TNF-α, as evidenced by neutralizing anti-TNF-α mAb in the in vivo Matrigel plug model [169]. High molecular weight kininogen (HK) is a plasma protein that is cleaved by plasma kallikrein, leading to the production of a kinin-free derivative of HK (i.e., HKa). The formation of HKa results in the exposure of its domain 5 (D5). HKa or D5 inhibit EC migration and proliferation. The mAb C11C1, which prevents the binding of HK to ECs, inhibited angiogenesis in experimental inflammatory arthritis [170]. In vitro, HKa or D5 abrogated EC adhesion to vitronectin and fibrinogen, causing anoikis and apoptosis. According to these findings, uPAR acting as a HKa receptor was considered to form a signaling complex containing αvβ3 and α5β1 and their associated signaling intermediates. HKa competes with vitronectin to bind to uPAR and disrupts this complex. Consequently, the mAb C11C1 has been proposed as a novel therapeutic to control angiogenesis in inflammation [170].
Since discovering the RGD motif, multiple peptidic and non-peptidic RGD-based integrin ligands with various degrees of specificity have been developed. Despite their similarities, the integrins can recognize different RGD-containing ECM proteins and respond differently to the interaction with each one of them [162]. Molecular imaging through RGD-based ligands for the non-invasive assessment of angiogenesis is of great current interest [171].
68Ga-Aquibeprin and 68Ga-Avebetrin are two PET tracers that have been explored in the early detection of arthritis in CIA: 68Ga-Aquibeprin as an α5β1 integrin targeted trimeric pseudopeptide and 68Ga-Avebetrin as an αvβ3 targeted cyclo (RDGfK) trimer [39,172]. Although both tracers detected arthritis disease activity, the α5β1 targeting tracer uptake was identified earlier than the αvβ3 targeted tracer in RA development, possibly even before the development of clinical symptoms. Interestingly, through comparison of the intensity of the integrin PET signals over time and correlation with the clinical RA indicators (i.e., arthritis scores), it was noted that during the progression of arthritis in CIA, the α5β1-integrin was not only upregulated earlier than αvβ3, but increased consistently, and elevated to a higher expression level than αvβ3 at later stages. Furthermore, the selectivity and specificity of both tracers to their respective integrin targets were confirmed by blockade and cross-blockade experiments. Cross-blockade by the saturation of the non-target integrin for each radiotracer did not influence the PET signal intensity, indicating that the observed signal in PET scans is only due to the binding of the corresponding radiolabeled ligand [39].
The first-in-human study to investigate the use of integrin in molecular imaging for the detection of synovial angiogenesis in RA utilized a cyclic PEGylated RGD peptide chelated with NOTA and monitored the response to treatment in comparison to 18F-FDG [79]. In this study, high levels of αvβ3 integrin expression were observed in IHC analysis in the vascular ECs of patients with active RA. 68Ga-PRGD2 PET/CT provided a better reflection of the disease severity and changes in the clinical measures than 18F-FDG in response to the therapeutic interventions. Additionally, the uptake pattern of the joints was different using 68Ga-PRGD2 imaging, where it displayed a diffuse synovial involvement of the affected joints and tendon sheaths of patients with RA, while the accumulation of 68Ga-PRGD2 was confined to a specific diseased area in patients with OA. Finally, unlike 18F-FDG, the integrin tracer did not accumulate in the axillary lymph nodes [79].
It has previously been reported that mast cells are the primary source of two important proangiogenic factors, IL-8 and TNF-α, in the inflamed synovial tissue. GPI-induced arthritis relies strongly on the presence of mast cells, but is largely independent of T and B cells. The activation of mast cells in this arthritis model has been found to activate αvβ3 integrin during early angiogenesis and induce pericyte proliferation, which occurs during the differentiation of mature blood vessels. Angiogenesis quantification by in vivo measuring of the activated αvβ3 integrin using 18F–galacto-RGD and IHC studies corroborated this finding [81].
The therapeutic effects of bevacizumab (Avastin), a humanized mAb against VEGF and the first FDA approved drug for tumor angiogenesis, have been explored by 99mTc-3P4-RGD2 in a rat CIA model. The in vivo imaging studies using this SPECT tracer demonstrated reduced uptake after TNF and VEGF therapy. Of note, both treatments exhibited similar effects on amelioration of arthritis [87].
99mTc-(HYNIC(tricine/TPPTS)-3PRGD2) is another RGD-based tracer that has been subjected to in vitro and in vivo evaluation for RA in a rat arthritis model. The radiolabeled tracer had renal excretion and was partly metabolized via the hepatobiliary route. Tracer uptake in the arthritic ankles was observed from 1 h onward with continuous visualization of arthritic ankles up to 6 h. The uptake of arthritic ankles of the rats in both planar imaging and biodistribution correlated well with the severity of arthritis. The linear relationship was also observed between the arthritic ankle uptake and the integrin αvβ3 expression levels, suggesting the practicability of molecular imaging and the estimation of αvβ3 expression for a selection of patients who may benefit from anti-angiogenesis therapy [83].
An in vivo SPECT imaging study with three different targets (i.e., FAP (111In-28H1), macrophages (111In-anti-F4/80-A3-1), and integrin αvβ3 (111In-DOTA-E-(cRGDfK)2) was performed to monitor the response to anti-TNF therapy (etanercept) in arthritic mice. Even though the three tracers differed in their targets, mechanism, and pharmacokinetics, the effect of etanercept on the macroscopic/clinical arthritis score could be quantitatively measured and correlated with all of the tracer imaging profiles. However, the decreased target uptake by these radiotracers after treatment needs further confirmation [86]. Of note, the RGD peptide-based tracer had a shorter imaging acquisition time (at 1 h) compared to the other two tracers 111In-anti-F4/80-A3-1 at 24 h and 111In-28H1 at 48 h.
Aiming to develop a novel PET tracer with high PET signal in vivo, three RGD peptides conjugated with a multimeric bifunctional chelator (tris(hydroxypyridinone)), to incorporate more than one radioisotope, were evaluated in serum transfer-induced arthritis in mice. Based on in vitro uptake, in vivo biodistribution, and target tissue accumulation, one conjugate (68Ga(HP3-RGD3)) was selected for arthritis imaging in severe and mild arthritis. 68Ga(HP3-RGD3) did not show statistically significant differences in joint uptake between the healthy mice and mice with mild arthritis, but was able to visualize severe arthritis. The uptake of 68Ga(HP3-RGD3) in severe arthritis was comparable to the previously studied 111In-DOTA-E(cRGDfK)2 tracer in CIA [80].
99mTc-3PRGD2 (99mTc-maraciclatide) scintigraphy showed a significant increase in tracer uptake in the joints of arthritic rats versus the untreated controls, consistent with the expression levels of αvβ3 and CD31 being higher in rat arthritic joint tissue compared to the controls. Bevacizumab therapy ameliorated the arthritis severity and reduced the radiotracer uptake in affected joints. This tracer was brought forward for testing in a patient with active RA where it proved its applicability in the early detection of synovial angiogenesis in RA. In contrast, bone scanning using 99mTc-MDP failed to distinguish early RA joints from the healthy controls [84].
The 99mTc-maraciclatide uptake in the joints of RA patients and its correlation with power Doppler (PD)US has also recently been studied. Both the PDUS and radiolabeled tracer results were in agreement with thee angiogenic markers and disease activity, however, planar imaging was considered to have the advantage of providing quantitative whole-body information with a single acquisition and easier interpretation compared to US/MRI. Further investigation in a larger study in RA patients has therefore been proposed [85].
The molecular imaging of angiogenesis by 68Ga-NODAGA-RGDyk and 18F-FDG was investigated in a patient with squamous cell carcinoma to detect the primary tumor and metastatic cervical lymph nodes. Interestingly, the uptake was also observed in the bilateral joints (the shoulders, elbows, wrists, metacarpophalangeal, interphalangeal, and hip joints) of the same patient, indicating moderately active RA. Indeed, this patient had previously been diagnosed with RA. Both tracers showed the same level of uptake in the arthritic joints, but the RGD peptide-based tracer exhibited a higher target-to-nontarget ratio [82].
Recently, a new diphosphine chemical platform (BMA or (diphosphine, 2,3-bis(diphenylphosphino)maleic anhydride)) enabling simple, one-step, kit-based 99mTc-radiolabeling of receptor-targeted peptides, has been employed for the synthesis of a cyclic RGD peptide (Arg-Gly-Asp-DPhe-Lys (RGD)). The radiolabeled tracer compound 99mTcO2(DP-RGD)2 demonstrated a high metabolic stability and selective accumulation in the joints of arthritic mice, which was in accordance with the joint swelling. Unlike other existing RGD-based radiotracers investigated thus far in RA, this one step, kit-based protocol, together with its comparable ability for angiogenesis detection, seems to pave the way for its widespread application in future studies [88].

3.6. Cyclooxygenase Enzymes

Cyclooxygenases (COXs) are key enzymes in prostaglandin (PG) biosynthesis from arachidonic acid metabolism and constitute the main target for non-steroidal anti-inflammatory drugs (NSAIDs) [173,174]. Two distinct isoforms of COX are expressed in the synovium. COX-1 is constitutively expressed, particularly by synovial lining cells, with no difference in its expression between inflammatory and OA. COX-2 is expressed by ECs, mononuclear leukocytes, and fibroblasts, and its expression is increased in inflammatory arthritis compared to that in OA [118]. COX-2 enzyme activity is upregulated in the rheumatoid synovium, leading to elevated PG production [175]. Both Prostaglandin E1 and Prostaglandin E2 (PGE1 and PGE2) are potent mediators of angiogenesis [176]. Pro-inflammatory cytokines such as IL-1β and TNF-α are also implicated in the overexpression of COX-2 [177]. The blockade of COX-2 expression by RNA-interference in RA fibroblast like synoviocyte (FLS) markedly reduced the VEGF levels. Similarly, COX-2 inhibitors can inhibit VEGF expression in RA FLSs; in contrast, exogenous PGE2 disrupts this inhibitory effect [174]. Studies exploring the effect of COX inhibitors on arthritic animal models have established that part of their effectiveness is mediated by suppressing the VEGF signaling pathway [178,179,180,181].
Some non-steroidal anti-inflammatory drugs (NSAIDs) have been radiolabeled for monitoring COX-1/-2 in RA. Using a CIA rat model of arthritis, PET imaging for COX-1 and COX-2 was performed with 11C-ketoprofen before and after TNF-α treatment (etanercept). The 11C-ketoprofen PET images showed a time-dependent accumulation of the tracer in the inflamed joint according to the progression of arthritis, although there was a moderate correlation between the tracer uptake and the level of paw swelling. After treatment, along with the decrease in paw swelling, the joint uptake was also reduced [90], underscoring its ability to monitor the inflammatory status during treatment. On the other hand, the development of a PET tracer by radiolabeling two isomers of ibuprofen (the (S)-1 isomer inhibits COX activity and (R)-1 isomer is inactive) with carbon-11 showed an increased accumulation of both radiotracers in the arthritic joints of animals. Lack of a different accumulation level between the enantiomers indicates that ibuprofen retention is regardless of the expression of COX expression [89].
Another novel PET tracer selectively targeting COX-2, 11C-MC1, was capable of visualizing the actively inflamed joints of RA patients in accordance with the increased number and activity of leukocytes in the synovial membranes. A blocking study with celecoxib, a preferential COX-2 inhibitor, confirmed the specificity of 11C-MC1 binding to COX-2 [91].

3.7. Cytokines

Pro-inflammatory cytokines may exert either direct angiogenic activity or indirect activity via the VEGF-dependent pathways [16]. TNF-α is a pleiotropic cytokine exerting diverse functions: the activation of leukocytes, ECs, and synovial fibroblasts; the induction of cytokines, chemokines, adhesion molecules, and matrix enzymes; the suppression of regulatory T-cell function; the activation of osteoclasts and the resorption of cartilage and bone; and the mediation of metabolic and cognitive dysfunction [182]. TNF-α is also considered as a potent pro-angiogenic mediator with an efficacy comparable to fibroblast growth factors (FGFs) [179]. TNF-α itself promotes neovascularization and may also regulate capillary formation via the angiopoietin 1 and angiopoietin 2 (Ang1/Ang2) Tie2-VEGF network [16,178,183]. In CIA, TNF-α regulated Tie2 activation involving interactions between ECs and synoviocytes. TNF-α upregulated Tie2 expression in ECs in a nuclear factor kappa B dependent process and also upregulated Ang1 in synoviocytes [184,185]. TNF-α stimulates the production of MMPs and PGE2 through the activation of intracellular signaling pathways including mitogen-activated protein kinase (MAPK) and activator protein-1(AP-1) [186]. The TNF-α blockade by infliximab, administered as a single agent or in combination with methotrexate, reduced the VEGF expression and vascularity within the RA synovium [16,187,188]. Simultaneous targeting of the TNF and Ang2 with a bispecific antibody showed anti-arthritic efficacy in an in vivo model of arthritis [189]. In an ex vivo setting, neo-angiogenesis in rheumatoid synovitis resulted from, at least in part, the angiogenic effect of locally produced TNF-α and platelet-activating factor (PAF) [190]. Consistently, (pre)clinical TNF blocking therapy studies reduced the joint inflammation by decreasing the expression of adhesion molecules as key players in the angiogenesis process [191,192,193,194,195]. Moreover, several novel natural and synthetic drugs such as triptolide, evening primrose oil (EPO) rich in gamma linolenic acid (GLA), niclosamide, wen luo yin, etc. have been shown to inhibit TNF-α production along with the inhibition of angiogenesis in vitro and in vivo in arthritis models [192,196,197,198,199].
Some studies have evaluated TNF-α expression using radiolabeled monoclonal antibodies (e.g., adalimumab and infliximab) or antibody fragments (e.g., certolizumab pegol) to evaluate the inflammatory activity in rheumatoid arthritis.
Infliximab, a chimeric anti-TNF-α antibody first used for RA treatment, was coupled with 99mTc and used to differentiate the responder to non-responder patients of anti-TNF-α therapy. The image analysis showed a trend of faster blood clearance due to the formation of infliximab–anti-infliximab complexes and higher liver/spleen uptake by the removal of the antibody complexes in a non-responding patient. Clinically inflamed joints also showed tracer uptake. Comparatively, another study using the same tracer confirmed the predictive role of 99mTc-anti-TNF mAb in therapy decision-making for the selection of RA candidates for infliximab therapy. In contrast, radiolabeled human nonspecific immunoglobulin (HIG) was not able to distinguish the changes in disease activity [96,97].
Adalimumab (Humira), a fully human, immunoglobulin G1 (IgG1), anti-TNF mAbm has also been assessed as an imaging modality [200] that could confirm or exclude RA diagnosis and select patients with disease activity to initiate anti-TNF-α therapy. In one study, the concordance of MRI for RA detection and 99mTc-adalimumab scintigraphy was strong. Furthermore, its performance in distinguishing between the inflammatory and non-inflammatory sites in the affected joints was reported to be of high accuracy and sensitivity, with specificity values around 90% [95].
Certolizumab pegol (CZP), another humanized mAb with a specificity for human TNF-α, has been investigated in different studies with different radiolabeling techniques for the imaging of patients with various types of inflammatory arthritis including RA. CZP differs from other TNF blockers due to its link with polyethylene glycol (PEG), which enhances the preferential distribution to inflamed tissue over normal tissue to a greater extent compared to adalimumab and infliximab [94,201]. These studies demonstrated the feasibility of immuno-PET imaging of CZP, which was radiolabeled with 89Zr or 99mTc for the visualization of TNF-driven diseases in a preclinical setting or in patients with active RA. Blocking tests confirmed that the PET images corresponded to the localization of active clinical inflammation and the tracers’ specificity [92,93,94].

3.8. Other

3.8.1. Microvascular Endothelium

A synovial endothelium targeting peptide (SyETP, CKSTHDRLC) has been identified to differentially locate at synovial xenografts in a human/SCID mouse chimeric model of RA, but not to the skin or mouse microvascular endothelium [202]. This peptide sequence maintained its affinity and specificity in vivo to bind to synovial microvascular ECs and has been proposed for use as a targeting carrier for selective drug delivery [203]. To this end, a (radiolabeled) novel fusion protein consisting of human IL-4 (anti-inflammatory cytokine) linked via a MMP-cleavable sequence to multiple copies of SyETP was evaluated for its anti-angiogenic potential in an in vivo SCID mouse model of arthritis along with imaging using nano-SPECT/CT. The iodine-125 labeled IL-4-SyETP accumulated in the synovial but not in the control skin xenografts [98].

3.8.2. Discussion

Synovial inflammation, particularly the proliferative pannus in RA, relies on synovial angiogenesis, which fuels multiple processes contributing to sustained inflammation in the course of the disease, and major pro-angiogenic factors have been identified in synovial fluid, synovial tissue, and plasma/serum [204]. Despite the discovery of angiogenesis as a key driver in RA pathobiology [205] and the identification of a large number of vascular targets, its utility for diagnostic non-invasive PET or SPECT imaging and therapeutics is still in its infancy. The majority of RA-related studies conducted in this context were performed at a preclinical level and should now proceed to the clinical assessment of the synovial neo-vasculature and its response to treatment. The molecular imaging studies targeting angiogenesis in RA have mostly been compared with 18F-FDG PET and bone scan (MDP/HDP) as reference molecular imaging tools. However, since these radiotracers lack specificity for the angiogenic process, there is an unmet need to identify more superior tracers for this purpose.
The six most promising tracers for the imaging of angiogenesis in RA, as described in the reported clinical studies, target binding sites in the ECM or on vascular adhesion molecules such as VAP-1 and αvβ3 and α5β1 integrins, which are upregulated in response to inflammation and growth factors. VAP-1 is a protein with a dual function as it acts both as an adhesion molecule and has amine oxidase enzymatic activity. Recently, the first in-human study with 68Ga-labeled siglec-9 as the VAP-1 ligand supported its candidature as a highly promising target to assess the angiogenic/inflammatory process in RA [73]. Other recent insights of sialylated siglec-9 being expressed on macrophages may add another role of VAP-1 in RA inflammatory programs [206].
Many efforts in integrin targeting have focused on αvβ3 with all radio-ligands applied in human RA studies being RGD-based peptides. The small size of the peptides allows them to be rapidly distributed and excreted from the body. Several promising variants of these RGD peptides with improved biodistribution and pharmacokinetics for αvβ3 integrin [79,84,207] are currently under clinical investigation for RA. Although αvβ3 is the main integrin used for imaging and targeting purposes, with relatively successful clinical application in the context of oncology and even in RA, there is still room for exploring other members of the integrin family. In this respect, ongoing screening studies have identified α5β1-selective novel (bicyclic) RGD peptides for future evaluation [162,208,209].
Synovial hypoxia is a hallmark triggering angiogenesis and five tracers (three for PET, two for SPECT) showed the feasibility of visualizing this process in RA already in the early stages of the disease. The chemical properties of the tracers accounted for a higher target to background ratio for 18F-FAZA (hydrophilic nature) than 18F-FMISO (lipophilic nature), which may determine the best choice for hypoxia imaging in RA for one of these or alternatives such as 64/67Cu-ATSM [210]. Of future interest, recent developments in hypoxia imaging in oncology include the testing of galectin tracers. Galectins, particularly galectin-1, -3, and-9, are implicated in angiogenesis [211,212,213,214] and in recent research, 68Ga-galectracer was evaluated for the PET imaging of tumor hypoxia, displaying superiority over FMISO in the imaging of hypoxia in animal models and allowed for the early prediction of the tumor response to radiotherapy [215]. Thus, 68Ga-galectracer may also be a candidate for imaging hypoxia in RA.
Diagnostic relevance of the somatostatin receptor has been established in different types of inflammatory diseases including RA with SST2 overexpression by ECs [216]. Initial scintigraphy studies with somatostatin analogue tracers claimed differential tracer binding to vascular pannus in active rheumatoid joints over non-vascularized joints [67,110], but since a very recent study with a somatostatin analogue PET tracer (DOTANOC) did not show added value over the reference MDP scan to detect clinically positive joints for RA, the clinical utility of somatostatin tracers warrants further exploration [65].
PET or SPECT imaging of MMPs is still in a preliminary phase both in oncology and RA studies, with no clinical evaluation reported to date [46]. Recently, potent MMP-13 inhibitors have been successfully synthesized and radiolabeled with the PET radioisotope. Further modifications to the tracers were recommended to improve their pharmacokinetic properties, after which (pre)clinical arthritis models are awaited to reveal their potential for angiogenesis imaging in RA.
Dual (low-dose) tracer imaging is another emerging strategy that can improve the contrast and specificity by providing a larger potential of binding sites, and as such, may be beneficial in the field of RA angiogenesis when it is hard to define and employ a single target for imaging. The conjugate F8-IL10 showed initial encouraging imaging results in RA animal and human studies, but awaits further confirmation. Another candidate as a dual targeting tracer for angiogenesis may be 68Ga-NOTA-3P-TATE-RGD, which binds specifically to integrin and somatostain [217,218]. Following oncology, this tracer is eligible for evaluating the RA angiogenesis in preclinical models.
Not all of the reported tracers in this overview specifically target RA angiogenesis. This holds relevance when evaluating the treatment efficacy of a compound related to its angiogenesis inhibitory effects. COX and TNF-α inhibitors are regularly used in RA clinical practice to elicit anti-inflammatory effects, but they also confer the inhibition of EC migration and synovial angiogenesis [16]. Their associated tracers may be particularly helpful in identifying patients responding to these therapies, although both the imaging and the clinical effects of these treatments may be largely independent of angiogenesis.
Angiogenesis is an attractive target due to its integral role in various diseases. Given the overlapping mechanisms between tumor and RA angiogenesis, potentially useful tracers identified in oncology in terms of desirable specificity, pharmacokinetics, and targeting opportunities support the translation of encouraging research outcomes to the RA field. Beyond the examples indicated above, some new angiogenic targets derived from oncology studies include urokinase plasminogen activator receptor (uPAR). uPAR on the neo-angiogenic endothelium supports the infiltration of inflammatory cells. 64Cu/68Ga-DOTA-AE105 is a peptide-based uPAR tracer in the clinical phase of testing, which may also be considered for the diagnosis of RA angiogenesis activity in the early phase of the disease [219]. The sphingosine-1-phosphate (S1P) pathway may also be another interesting target to investigate in the context of angiogenesis therapy and imaging. This target is among a growing number of studies that have investigated its role in RA and malignancies [7,220,221,222,223,224,225]. Imaging modalities are becoming increasingly important for diagnostic and therapy response monitoring studies. Conceivably, imaging can also be positioned in areas such as the stratification of patients for personalized medicine treatment, prognosis and pathogenetic research. As such, angiogenesis imaging can contribute, to a certain degree, to the optimal diagnosis and treatment of RA patients.
The current review had some limitations. The parameters in the search strategy were chosen to find the angiogenic targets involved in RA and based on those to identify the molecular imaging markers (tracers) that have the potential to visualize angiogenesis in RA in clinical and preclinical phases of the disease. This search strategy could have led to some relevant studies being missed, especially in the preclinical stages of the disease as well as radiopharmaceuticals that have not been primarily tested or developed for RA. In addition, angiogenesis biomarkers possess multiple functions in RA and thus may have been missed if described under an alternative role. Finally, the restriction of the search strategy with three terms to identify angiogenesis targets could also have resulted in missing applicable targets.

4. Conclusions

This review provided a summary of the molecular imaging tracers with a potential applicability in the detection or monitoring of angiogenic activities as an important component of RA disease. Adhesion molecules particularly targeting VAP-1 and integrins, up to now, seem to hold promising results due to their critical roles in angiogenesis. Nonetheless, to identify the most appropriate angiogenic molecular target for imaging, further performance optimizations and/or clinical evaluations of the currently introduced radiotracers as well as the exploration of new targets are required to expand our insights into exploiting angiogenesis in RA. Overall, targeting angiogenesis by radiotracers holds great promise for the early diagnosis, selection of patients, and treatment evaluation in RA.

Supplementary Materials

The following supporting information can be downloaded at: https://www.mdpi.com/article/10.3390/ijms23137071/s1, File S1. Specification of RA angiogenesis biomarkers search_March 2022. File S2. Specification of RA radiotracers search_March 2022.

Author Contributions

Writing—original draft preparation, F.K.; Figure design, F.K.; Review and editing, A.E., M.M.S., J.R.v.B., G.J.C.Z., G.J., S.W.T. and C.J.v.d.L. All authors have read and agreed to the published version of the manuscript.

Funding

This project received funding from the European Union’s Horizon 2020 research and innovation program under the Marie Skłodowska-Curie Grant Agreement No. 847551.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We would like to thank the medical information specialist, Ralph de Vries, Medical Library, Vrije Universiteit, Amsterdam, The Netherlands, for his expert help in optimizing the search strategies applied in this manuscript as well as Merve Kocygit (M.K.), Amsterdam UMC, Amsterdam, The Netherlands, for her contribution in screening and extracting the data for this review. All illustrations were created as original figures specific for this review with BioRender.com (accessed on 23 November 2021).

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

Ang1/2Angiopoietin 1/2
A-SSAAcute-phase serum amyloid A
CAMCell adhesion molecules
CIACollagen-induced arthritis
COXCyclooxygenases
CTComputed tomography
Cu-ATSMCopper(II) diacetyl-di(N4-methylthiosemicarbazone)
DMARDDisease-modifying anti-rheumatic drug
DOTA2,2′,2″,2‴-(1,4,7,10-Tetraazacyclododecane-1,4,7,10-tetrayl)tetraacetic acid OR tetraxetan
ECMExtracellular matrix
ED-A/-BExtra domain of fibronectin-A/-B
FAPFibroblast activation protein
FAZAFluoroazomycinarabinoside
FDGFluorodeoxyglucose
FLSFibroblast-like synoviocyte
FMISOFluoromisonidazole)
HDPHydroxydiphosphonate
HIGHuman immunoglobulin
HUVECHuman umbilical vein endothelial cell
HYNICHydrazinonicotinamide
IAZA1-(α-D-5-Iodoarabinofuranosyl)-2-nitroimidazole)
IHCImmunohistochemistry
ILInterleukin
JAKJanus kinase
mAbMonoclonal antibody
MAPKMitogen-activated protein kinase
MDPMethyl diphosphonate
MMPMatrix metalloproteinases
MRIMagnetic resonance imaging
MT-MMPsMembrane-type matrix metalloproteinases
NOTA1,4,7-Triazacyclononane-1,4,7-triacetic acid
OAOsteoarthritis
GPIGlucose-6-phosphate isomerase
PDUSPower Doppler ultra-sonography
PEGPoly(ethylene glycol)
PETPositron emission tomography
PGEProstaglandin E
PI3KPhosphoinositide 3-kinases
RARheumatoid arthritis
RGDArginylglycylaspartic acid
SCIDSevere combined immunodeficiency disease
SPECTPhoton emission computed tomography
SRCSRC proto-oncogene, non-receptor tyrosine kinase
SSTSomatostatin
STATSignal transducer and activator of transcription
TNFTumor necrosis factor
TRAPTriazacyclononane-triphosphinate
uPARUrokinase plasminogen activator receptor
VAP-1Vascular adhesion protein
VEGFRVascular endothelial growth factor receptor

References

  1. Smolen, J.S.; Aletaha, D.; Barton, A.; Burmester, G.R.; Emery, P.; Firestein, G.S.; Kavanaugh, A.; McInnes, I.B.; Solomon, D.H.; Strand, V.; et al. Rheumatoid arthritis. Nat. Rev. Dis Primers 2018, 4, 18001. [Google Scholar] [CrossRef] [PubMed]
  2. Guo, Q.; Wang, Y.; Xu, D.; Nossent, J.; Pavlos, N.J.; Xu, J. Rheumatoid arthritis: Pathological mechanisms and modern pharmacologic therapies. Bone Res. 2018, 6, 15. [Google Scholar] [CrossRef] [PubMed]
  3. Leblond, A.; Allanore, Y.; Avouac, J. Targeting synovial neoangiogenesis in rheumatoid arthritis. Autoimmun. Rev. 2017, 16, 594–601. [Google Scholar] [CrossRef] [PubMed]
  4. Kim, H.R.; Kim, K.W.; Kim, B.M.; Cho, M.L.; Lee, S.H. The effect of vascular endothelial growth factor on osteoclastogenesis in rheumatoid arthritis. PLoS ONE 2015, 10, e0124909. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Orr, C.; Vieira-Sousa, E.; Boyle, D.L.; Buch, M.H.; Buckley, C.D.; Canete, J.D.; Catrina, A.I.; Choy, E.H.S.; Emery, P.; Fearon, U.; et al. Synovial tissue research: A state-of-the-art review. Nat. Rev. Rheumatol. 2017, 13, 463–475. [Google Scholar] [CrossRef]
  6. Haubner, R.; Beer, A.J.; Wang, H.; Chen, X. Positron emission tomography tracers for imaging angiogenesis. Eur. J. Nucl. Med. Mol. Imaging 2010, 37, 86–103. [Google Scholar] [CrossRef] [Green Version]
  7. Tas, S.W.; Maracle, C.X.; Balogh, E.; Szekanecz, Z. Targeting of proangiogenic signalling pathways in chronic inflammation. Nat. Rev. Rheumatol. 2016, 12, 111–122. [Google Scholar] [CrossRef]
  8. Vailhe, B.; Vittet, D.; Feige, J.J. In vitro models of vasculogenesis and angiogenesis. Lab. Investig. 2001, 81, 439–452. [Google Scholar] [CrossRef] [Green Version]
  9. Patan, S. Vasculogenesis and angiogenesis. Cancer Treat Res. 2004, 117, 3–32. [Google Scholar]
  10. Elshabrawy, H.A.; Chen, Z.; Volin, M.V.; Ravella, S.; Virupannavar, S.; Shahrara, S. The pathogenic role of angiogenesis in rheumatoid arthritis. Angiogenesis 2015, 18, 433–448. [Google Scholar] [CrossRef] [Green Version]
  11. Wang, Y.; Wu, H.; Deng, R. Angiogenesis as a potential treatment strategy for rheumatoid arthritis. Eur. J. Pharmacol. 2021, 910, 174500. [Google Scholar] [CrossRef] [PubMed]
  12. George, G.; Shyni, G.L.; Raghu, K.G. Current and novel therapeutic targets in the treatment of rheumatoid arthritis. Inflammopharmacology 2020, 28, 1457–1476. [Google Scholar] [CrossRef] [PubMed]
  13. Griffioen, A.W.; Molema, G. Angiogenesis: Potentials for pharmacologic intervention in the treatment of cancer, cardiovascular diseases, and chronic inflammation. Pharmacol. Rev. 2000, 52, 237–268. [Google Scholar] [PubMed]
  14. Chen, Q.; Fan, K.; Chen, X.; Xie, X.; Huang, L.; Song, G.; Qi, W. Ezrin regulates synovial angiogenesis in rheumatoid arthritis through YAP and Akt signalling. J. Cell Mol. Med. 2021, 25, 9378–9389. [Google Scholar] [CrossRef] [PubMed]
  15. Florea, A.; Mottaghy, F.M.; Bauwens, M. Molecular Imaging of Angiogenesis in Oncology: Current Preclinical and Clinical Status. Int J. Mol. Sci 2021, 22, 5544. [Google Scholar] [CrossRef] [PubMed]
  16. Szekanecz, Z.; Besenyei, T.; Paragh, G.; Koch, A.E. Angiogenesis in rheumatoid arthritis. Autoimmunity 2009, 42, 563–573. [Google Scholar] [CrossRef] [Green Version]
  17. Peluzzo, A.M.; Autieri, M.V. Challenging the Paradigm: Anti-Inflammatory Interleukins and Angiogenesis. Cells 2022, 11, 587. [Google Scholar] [CrossRef]
  18. Clavel, G.; Bessis, N.; Lemeiter, D.; Fardellone, P.; Mejjad, O.; Menard, J.F.; Pouplin, S.; Boumier, P.; Vittecoq, O.; Le Loet, X.; et al. Angiogenesis markers (VEGF, soluble receptor of VEGF and angiopoietin-1) in very early arthritis and their association with inflammation and joint destruction. Clin. Immunol. 2007, 124, 158–164. [Google Scholar] [CrossRef]
  19. Lee, S.S.; Joo, Y.S.; Kim, W.U.; Min, D.J.; Min, J.K.; Park, S.H.; Cho, C.S.; Kim, H.Y. Vascular endothelial growth factor levels in the serum and synovial fluid of patients with rheumatoid arthritis. Clin. Exp. Rheumatol. 2001, 19, 321–324. [Google Scholar]
  20. Ozgonenel, L.; Cetin, E.; Tutun, S.; Tonbaklar, P.; Aral, H.; Guvenen, G. The relation of serum vascular endothelial growth factor level with disease duration and activity in patients with rheumatoid arthritis. Clin. Rheumatol. 2010, 29, 473–477. [Google Scholar] [CrossRef]
  21. Harada, M.; Mitsuyama, K.; Yoshida, H.; Sakisaka, S.; Taniguchi, E.; Kawaguchi, T.; Ariyoshi, M.; Saiki, T.; Sakamoto, M.; Nagata, K.; et al. Vascular endothelial growth factor in patients with rheumatoid arthritis. Scand. J. Rheumatol. 1998, 27, 377–380. [Google Scholar] [PubMed]
  22. Sone, H.; Sakauchi, M.; Takahashi, A.; Suzuki, H.; Inoue, N.; Iida, K.; Shimano, H.; Toyoshima, H.; Kawakami, Y.; Okuda, Y.; et al. Elevated levels of vascular endothelial growth factor in the sera of patients with rheumatoid arthritis correlation with disease activity. Life Sci. 2001, 69, 1861–1869. [Google Scholar] [CrossRef]
  23. Leblond, A.; Pezet, S.; Trouvin, A.P.; Elhai, M.; Gonzalez, V.; Allanore, Y.; Avouac, J. Linking systemic angiogenic markers to synovial vascularization in rheumatoid arthritis. PLoS ONE 2018, 13, e0203607. [Google Scholar] [CrossRef]
  24. Kim, J.W.; Kong, J.S.; Lee, S.; Yoo, S.A.; Koh, J.H.; Jin, J.; Kim, W.U. Angiogenic cytokines can reflect the synovitis severity and treatment response to biologics in rheumatoid arthritis. Exp. Mol. Med. 2020, 52, 843–853. [Google Scholar] [CrossRef] [PubMed]
  25. Nagashima, M.; Wauke, K.; Hirano, D.; Ishigami, S.; Aono, H.; Takai, M.; Sasano, M.; Yoshino, S. Effects of combinations of anti-rheumatic drugs on the production of vascular endothelial growth factor and basic fibroblast growth factor in cultured synoviocytes and patients with rheumatoid arthritis. Rheumatology 2000, 39, 1255–1262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Paleolog, E.M.; Young, S.; Stark, A.C.; McCloskey, R.V.; Feldmann, M.; Maini, R.N. Modulation of angiogenic vascular endothelial growth factor by tumor necrosis factor alpha and interleukin-1 in rheumatoid arthritis. Arthritis Rheum. 1998, 41, 1258–1265. [Google Scholar] [CrossRef]
  27. Nagashima, M.; Yoshino, S.; Aono, H.; Takai, M.; Sasano, M. Inhibitory effects of anti-rheumatic drugs on vascular endothelial growth factor in cultured rheumatoid synovial cells. Clin. Exp. Immunol. 1999, 116, 360–365. [Google Scholar] [CrossRef]
  28. Macias, I.; Garcia-Perez, S.; Ruiz-Tudela, M.; Medina, F.; Chozas, N.; Giron-Gonzalez, J.A. Modification of pro- and antiinflammatory cytokines and vascular-related molecules by tumor necrosis factor-a blockade in patients with rheumatoid arthritis. J. Rheumatol. 2005, 32, 2102–2108. [Google Scholar]
  29. Umemura, M.; Isozaki, T.; Ishii, S.; Seki, S.; Oguro, N.; Miura, Y.; Miwa, Y.; Nakamura, M.; Inagaki, K.; Kasama, T. Reduction of Serum ADAM17 Level Accompanied with Decreased Cytokines after Abatacept Therapy in Patients with Rheumatoid Arthritis. Int. J. Biomed. Sci. 2014, 10, 229–235. [Google Scholar]
  30. Hirohata, S.; Abe, A.; Murasawa, A.; Kanamono, T.; Tomita, T.; Yoshikawa, H. Differential effects of IL-6 blockade tocilizumab and TNF inhibitors on angiogenesis in synovial tissues from patients with rheumatoid arthritis. Mod. Rheumatol. 2017, 27, 766–772. [Google Scholar] [CrossRef]
  31. Di Benedetto, P.; Ruscitti, P.; Berardicurti, O.; Panzera, N.; Grazia, N.; Di Vito Nolfi, M.; Di Francesco, B.; Navarini, L.; Maurizi, A.; Rucci, N.; et al. Blocking Jak/STAT signalling using tofacitinib inhibits angiogenesis in experimental arthritis. Arthritis Res. Ther. 2021, 23, 213. [Google Scholar] [CrossRef] [PubMed]
  32. Zisman, D.; Safieh, M.; Simanovich, E.; Feld, J.; Kinarty, A.; Zisman, L.; Gazitt, T.; Haddad, A.; Elias, M.; Rosner, I.; et al. Tocilizumab (TCZ) Decreases Angiogenesis in Rheumatoid Arthritis Through Its Regulatory Effect on miR-146a-5p and EMMPRIN/CD147. Front. Immunol. 2021, 12, 739592. [Google Scholar] [CrossRef] [PubMed]
  33. Zhang, W.; Li, F.; Gao, W. Tripterygium wilfordii Inhibiting Angiogenesis for Rheumatoid Arthritis Treatment. J. Natl. Med. Assoc. 2017, 109, 142–148. [Google Scholar] [CrossRef]
  34. Bainbridge, J.; Sivakumar, B.; Paleolog, E. Angiogenesis as a therapeutic target in arthritis: Lessons from oncology. Curr. Pharm. Des. 2006, 12, 2631–2644. [Google Scholar] [CrossRef] [PubMed]
  35. Tan, Y.K.; Conaghan, P.G. Imaging in rheumatoid arthritis. Best Pract. Res. Clin. Rheumatol. 2011, 25, 569–584. [Google Scholar] [CrossRef] [PubMed]
  36. Scherer, H.U.; Haupl, T.; Burmester, G.R. The etiology of rheumatoid arthritis. J. Autoimmun. 2020, 110, 102400. [Google Scholar] [CrossRef] [PubMed]
  37. Aletaha, D.; Neogi, T.; Silman, A.J.; Funovits, J.; Felson, D.T.; Bingham, C.O., 3rd; Birnbaum, N.S.; Burmester, G.R.; Bykerk, V.P.; Cohen, M.D.; et al. 2010 rheumatoid arthritis classification criteria: An American College of Rheumatology/European League Against Rheumatism collaborative initiative. Ann. Rheum. Dis. 2010, 69, 1580–1588. [Google Scholar] [CrossRef] [PubMed]
  38. Kgoebane, K.; Ally, M.; Duim-Beytell, M.C.; Suleman, F.E. The role of imaging in rheumatoid arthritis. SA J. Radiol. 2018, 22, 1316. [Google Scholar] [CrossRef] [PubMed]
  39. Notni, J.; Gassert, F.T.; Steiger, K.; Sommer, P.; Weichert, W.; Rummeny, E.J.; Schwaiger, M.; Kessler, H.; Meier, R.; Kimm, M.A. In vivo imaging of early stages of rheumatoid arthritis by alpha5beta1-integrin-targeted positron emission tomography. EJNMMI Res. 2019, 9, 87. [Google Scholar] [CrossRef] [Green Version]
  40. Rosado-de-Castro, P.H.; Lopes de Souza, S.A.; Alexandre, D.; Barbosa da Fonseca, L.M.; Gutfilen, B. Rheumatoid arthritis: Nuclear Medicine state-of-the-art imaging. World J. Orthop. 2014, 5, 312. [Google Scholar] [CrossRef]
  41. van der Meulen, N.P.; Strobel, K.; Lima, T.V.M. New Radionuclides and Technological Advances in SPECT and PET Scanners. Cancers 2021, 13, 6183. [Google Scholar] [CrossRef] [PubMed]
  42. Stacy, M.R.; Maxfield, M.W.; Sinusas, A.J. Targeted molecular imaging of angiogenesis in PET and SPECT: A review. Yale J. Biol. Med. 2012, 85, 75–86. [Google Scholar] [PubMed]
  43. Shaw, R.C.; Tamagnan, G.D.; Tavares, A.A.S. Rapidly (and Successfully) Translating Novel Brain Radiotracers From Animal Research Into Clinical Use. Front. Neurosci. 2020, 14, 871. [Google Scholar] [CrossRef]
  44. Kostelnik, T.I.; Orvig, C. Radioactive Main Group and Rare Earth Metals for Imaging and Therapy. Chem. Rev. 2019, 119, 902–956. [Google Scholar] [CrossRef] [PubMed]
  45. Price, E.W.; Orvig, C. Matching chelators to radiometals for radiopharmaceuticals. Chem. Soc. Rev. 2014, 43, 260–290. [Google Scholar] [CrossRef] [PubMed]
  46. Hong, H.; Chen, F.; Zhang, Y.; Cai, W. New radiotracers for imaging of vascular targets in angiogenesis-related diseases. Adv. Drug Deliv. Rev. 2014, 76, 2–20. [Google Scholar] [CrossRef] [Green Version]
  47. Okoye, N.C.; Baumeister, J.E.; Khosroshahi, F.N.; Hennkens, H.M.; Jurisson, S.S. Chelators and metal complex stability for radiopharmaceutical applications. Radiochim. Acta 2019, 107, 1087–1120. [Google Scholar] [CrossRef]
  48. Evans, B.J.; King, A.T.; Katsifis, A.; Matesic, L.; Jamie, J.F. Methods to Enhance the Metabolic Stability of Peptide-Based PET Radiopharmaceuticals. Molecules 2020, 25, 2314. [Google Scholar] [CrossRef]
  49. Sharma, R.; Aboagye, E. Development of radiotracers for oncology--the interface with pharmacology. Br. J. Pharmacol. 2011, 163, 1565–1585. [Google Scholar] [CrossRef] [Green Version]
  50. Gotthardt, M.; Bleeker-Rovers, C.P.; Boerman, O.C.; Oyen, W.J. Imaging of inflammation by PET, conventional scintigraphy, and other imaging techniques. J. Nucl. Med. Technol. 2013, 41, 157–169. [Google Scholar] [CrossRef] [Green Version]
  51. Nadig, V.; Herrmann, K.; Mottaghy, F.M.; Schulz, V. Hybrid total-body pet scanners-current status and future perspectives. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 445–459. [Google Scholar] [CrossRef] [PubMed]
  52. Bowden, D.J.; Barrett, T. Angiogenesis imaging in neoplasia. J. Clin. Imaging Sci. 2011, 1, 38. [Google Scholar] [CrossRef] [PubMed]
  53. Niccoli Asabella, A.; Di Palo, A.; Altini, C.; Ferrari, C.; Rubini, G. Multimodality Imaging in Tumor Angiogenesis: Present Status and Perspectives. Int J. Mol. Sci. 2017, 18, 1864. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Pirovano, G.; Roberts, S.; Kossatz, S.; Reiner, T. Optical Imaging Modalities: Principles and Applications in Preclinical Research and Clinical Settings. J. Nucl. Med. 2020, 61, 1419–1427. [Google Scholar] [CrossRef] [PubMed]
  55. Pysz, M.A.; Gambhir, S.S.; Willmann, J.K. Molecular imaging: Current status and emerging strategies. Clin. Radiol. 2010, 65, 500–516. [Google Scholar] [CrossRef] [Green Version]
  56. Kubota, K.; Yamashita, H.; Mimori, A. Clinical Value of FDG-PET/CT for the Evaluation of Rheumatic Diseases: Rheumatoid Arthritis, Polymyalgia Rheumatica, and Relapsing Polychondritis. Semin. Nucl. Med. 2017, 47, 408–424. [Google Scholar] [CrossRef]
  57. Narayan, N.; Owen, D.R.; Taylor, P.C. Advances in positron emission tomography for the imaging of rheumatoid arthritis. Rheumatology 2017, 56, 1837–1846. [Google Scholar] [CrossRef] [Green Version]
  58. Page, M.J.; McKenzie, J.E.; Bossuyt, P.M.; Boutron, I.; Hoffmann, T.C.; Mulrow, C.D.; Shamseer, L.; Tetzlaff, J.M.; Akl, E.A.; Brennan, S.E.; et al. The PRISMA 2020 statement: An updated guideline for reporting systematic reviews. PLoS Med. 2021, 18, e1003583. [Google Scholar] [CrossRef]
  59. Ouzzani, M.; Hammady, H.; Fedorowicz, Z.; Elmagarmid, A. Rayyan-a web and mobile app for systematic reviews. Syst. Rev. 2016, 5, 210. [Google Scholar] [CrossRef] [Green Version]
  60. Guenthoer, P.; Fuchs, K.; Reischl, G.; Quintanilla-Martinez, L.; Gonzalez-Menendez, I.; Laufer, S.; Pichler, B.J.; Kneilling, M. Evaluation of the therapeutic potential of the selective p38 MAPK inhibitor Skepinone-L and the dual p38/JNK 3 inhibitor LN 950 in experimental K/BxN serum transfer arthritis. Inflammopharmacology 2019, 27, 1217–1227. [Google Scholar] [CrossRef]
  61. Fuchs, K.; Kuehn, A.; Mahling, M.; Guenthoer, P.; Hector, A.; Schwenck, J.; Hartl, D.; Laufer, S.; Kohlhofer, U.; Quintanilla-Martinez, L.; et al. In Vivo Hypoxia PET Imaging Quantifies the Severity of Arthritic Joint Inflammation in Line with Overexpression of Hypoxia-Inducible Factor and Enhanced Reactive Oxygen Species Generation. J. Nucl. Med. 2017, 58, 853–860. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Dam, T.T.; Hanaoka, H.; Nakajima, T.; Yamaguchi, A.; Okamura, K.; Chikuda, H.; Tsushima, Y. (64)Cu-ATSM and (99m)Tc(CO)3-DCM20 potential in the early detection of rheumatoid arthritis. Mod. Rheumatol. 2021, 31, 350–356. [Google Scholar] [CrossRef] [PubMed]
  63. Kumar, P.; Wiebe, L.I.; Atrazheva, E.; Tandon, M. An improved synthesis of a-AZA, a-AZP and a-AZG, the precursors to clinical markers of tissue hypoxia. Tetrahedron Lett. 2001, 42, 2077–2088. [Google Scholar] [CrossRef]
  64. Wiebe, I.L.; McEwan, A.J. Scintigraphic imaging of focal hypoxic tissue: Development and clinical applications of 123IAZA. Braz. Arch. Biol. Techology 2002, 45, 69–81. [Google Scholar] [CrossRef]
  65. Shamim, S.A.; Arora, G.; Kumar, N.; Behera, A.; Hussain, J.; Gupta, R.; Kumar, R.; Bal, C. Comparison of 99mTc-methyl diphosphonate bone scintigraphy and 68Ga-DOTANOC PET/computed tomography in articular manifestation of rheumatoid arthritis. Nucl. Med. Commun. 2022, 43, 428–432. [Google Scholar] [CrossRef]
  66. Anzola-Fuentes, L.K.; Chianelli, M.; Galli, F.; Glaudemans, A.W.; Martin Martin, L.; Todino, V.; Migliore, A.; Signore, A. Somatostatin receptor scintigraphy in patients with rheumatoid arthritis and secondary Sjogren’s syndrome treated with Infliximab: A pilot study. EJNMMI Res. 2016, 6, 49. [Google Scholar] [CrossRef] [Green Version]
  67. Vanhagen, P.M.; Markusse, H.M.; Lamberts, S.W.; Kwekkeboom, D.J.; Reubi, J.C.; Krenning, E.P. Somatostatin receptor imaging. The presence of somatostatin receptors in rheumatoid arthritis. Arthritis Rheum. 1994, 37, 1521–1527. [Google Scholar] [CrossRef] [Green Version]
  68. Papathanasiou, N.D.; Rondogianni, P.E.; Pianou, N.K.; Karampina, P.A.; Vlontzou, E.A.; Datseris, I.E. 99mTc-depreotide in the evaluation of bone infection and inflammation. Nucl. Med. Commun. 2008, 29, 239–246. [Google Scholar] [CrossRef]
  69. Schrigten, D.; Breyholz, H.J.; Wagner, S.; Hermann, S.; Schober, O.; Schafers, M.; Haufe, G.; Kopka, K. A new generation of radiofluorinated pyrimidine-2,4,6-triones as MMP-targeted radiotracers for positron emission tomography. J. Med. Chem. 2012, 55, 223–232. [Google Scholar] [CrossRef]
  70. Hugenberg, V.; Wagner, S.; Kopka, K.; Schafers, M.; Schuit, R.C.; Windhorst, A.D.; Hermann, S. Radiolabeled Selective Matrix Metalloproteinase 13 (MMP-13) Inhibitors: (Radio)Syntheses and in Vitro and First in Vivo Evaluation. J. Med. Chem. 2017, 60, 307–321. [Google Scholar] [CrossRef] [Green Version]
  71. Rangasamy, L.; Geronimo, B.D.; Ortin, I.; Coderch, C.; Zapico, J.M.; Ramos, A.; de Pascual-Teresa, B. Molecular Imaging Probes Based on Matrix Metalloproteinase Inhibitors (MMPIs). Molecules 2019, 24, 2982. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Bruijnen, S.T.G.; Chandrupatla, D.; Giovanonni, L.; Neri, D.; Vugts, D.J.; Huisman, M.C.; Hoekstra, O.S.; Musters, R.J.P.; Lammertsma, A.A.; van Dongen, G.; et al. F8-IL10: A New Potential Antirheumatic Drug Evaluated by a PET-Guided Translational Approach. Mol. Pharm. 2019, 16, 273–281. [Google Scholar] [CrossRef] [PubMed]
  73. Viitanen, R.; Moisio, O.; Lankinen, P.; Li, X.G.; Koivumaki, M.; Suilamo, S.; Tolvanen, T.; Taimen, K.; Mali, M.; Kohonen, I.; et al. First-in-Humans Study of (68)Ga-DOTA-Siglec-9, a PET Ligand Targeting Vascular Adhesion Protein 1. J. Nucl. Med. 2021, 62, 577–583. [Google Scholar] [CrossRef]
  74. Virtanen, H.; Autio, A.; Siitonen, R.; Liljenback, H.; Saanijoki, T.; Lankinen, P.; Makila, J.; Kakela, M.; Teuho, J.; Savisto, N.; et al. 68Ga-DOTA-Siglec-9--a new imaging tool to detect synovitis. Arthritis Res. Ther. 2015, 17, 308. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  75. Autio, A.; Vainio, P.J.; Suilamo, S.; Mali, A.; Vainio, J.; Saanijoki, T.; Noponen, T.; Ahtinen, H.; Luoto, P.; Teras, M.; et al. Preclinical evaluation of a radioiodinated fully human antibody for in vivo imaging of vascular adhesion protein-1-positive vasculature in inflammation. J. Nucl. Med. 2013, 54, 1315–1319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Garrood, T.; Blades, M.; Haskard, D.O.; Mather, S.; Pitzalis, C. A novel model for the pre-clinical imaging of inflamed human synovial vasculature. Rheumatology 2009, 48, 926–931. [Google Scholar] [CrossRef] [Green Version]
  77. Jamar, F.; Houssiau, F.A.; Devogelaer, J.P.; Chapman, P.T.; Haskard, D.O.; Beaujean, V.; Beckers, C.; Manicourt, D.H.; Peters, A.M. Scintigraphy using a technetium 99m-labelled anti-E-selectin Fab fragment in rheumatoid arthritis. Rheumatology 2002, 41, 53–61. [Google Scholar] [CrossRef] [Green Version]
  78. Jamar, F.; Chapman, P.T.; Manicourt, D.H.; Glass, D.M.; Haskard, D.O.; Peters, A.M. A comparison between 111In-anti-E-selectin mAb and 99Tcm-labelled human non-specific immunoglobulin in radionuclide imaging of rheumatoid arthritis. Br. J. Radiol. 1997, 70, 473–481. [Google Scholar] [CrossRef]
  79. Zhu, Z.; Yin, Y.; Zheng, K.; Li, F.; Chen, X.; Zhang, F.; Zhang, X. Evaluation of synovial angiogenesis in patients with rheumatoid arthritis using (6)(8)Ga-PRGD2 PET/CT: A prospective proof-of-concept cohort study. Ann. Rheum. Dis. 2014, 73, 1269–1272. [Google Scholar] [CrossRef] [Green Version]
  80. Imberti, C.; Terry, S.Y.; Cullinane, C.; Clarke, F.; Cornish, G.H.; Ramakrishnan, N.K.; Roselt, P.; Cope, A.P.; Hicks, R.J.; Blower, P.J.; et al. Enhancing PET Signal at Target Tissue in Vivo: Dendritic and Multimeric Tris(hydroxypyridinone) Conjugates for Molecular Imaging of alphavbeta3 Integrin Expression with Gallium-68. Bioconjug. Chem. 2017, 28, 481–495. [Google Scholar] [CrossRef]
  81. Kneilling, M.; Hultner, L.; Pichler, B.J.; Mailhammer, R.; Morawietz, L.; Solomon, S.; Eichner, M.; Sabatino, J.; Biedermann, T.; Krenn, V.; et al. Targeted mast cell silencing protects against joint destruction and angiogenesis in experimental arthritis in mice. Arthritis Rheum. 2007, 56, 1806–1816. [Google Scholar] [CrossRef] [PubMed]
  82. Dietz, M.; Nicod Lalonde, M.; Omoumi, P.; Testart Dardel, N.; Hügle, T.; Prior, J.O. Imaging of ανβ3 integrin expression in rheumatoid arthritis with [68Ga]Ga-NODAGA-RGDyk PET/CT in comparison to [18F]FDG PET/CT. Med. Nucl. 2021, 45, 293–295. [Google Scholar] [CrossRef]
  83. Huang, C.; Zheng, Q.; Miao, W. Study of novel molecular probe 99mTc-3PRGD2 in the diagnosis of rheumatoid arthritis. Nucl. Med. Commun. 2015, 36, 1208–1214. [Google Scholar] [CrossRef] [PubMed]
  84. Wu, Y.; Zhang, G.; Wang, X.; Zhao, Z.; Wang, T.; Wang, X.; Li, X.F. Early detection of rheumatoid arthritis in rats and humans with 99mTc-3PRGD2 scintigraphy: Imaging synovial neoangiogenesis. Oncotarget 2017, 8, 5753–5760. [Google Scholar] [CrossRef] [Green Version]
  85. Attipoe, L.; Chaabo, K.; Wajed, J.; Hassan, F.U.; Shivapatham, D.; Morrison, M.; Ballinger, J.; Cook, G.; Cope, A.P.; Garrood, T. Imaging neoangiogenesis in rheumatoid arthritis (INIRA): Whole-body synovial uptake of a (99m)Tc-labelled RGD peptide is highly correlated with power Doppler ultrasound. Ann. Rheum. Dis. 2020, 79, 1254–1255. [Google Scholar] [CrossRef]
  86. Terry, S.Y.; Koenders, M.I.; Franssen, G.M.; Nayak, T.K.; Freimoser-Grundschober, A.; Klein, C.; Oyen, W.J.; Boerman, O.C.; Laverman, P. Monitoring Therapy Response of Experimental Arthritis with Radiolabeled Tracers Targeting Fibroblasts, Macrophages, or Integrin alphavbeta3. J. Nucl. Med. 2016, 57, 467–472. [Google Scholar] [CrossRef] [Green Version]
  87. Wang, Y.; Da, G.; Li, H.; Zheng, Y. Avastin exhibits therapeutic effects on collagen-induced arthritis in rat model. Inflammation 2013, 36, 1460–1467. [Google Scholar] [CrossRef]
  88. Hungnes, I.N.; Al-Salemee, F.; Gawne, P.J.; Eykyn, T.; Atkinson, R.A.; Terry, S.Y.A.; Clarke, F.; Blower, P.J.; Pringle, P.G.; Ma, M.T. One-step, kit-based radiopharmaceuticals for molecular SPECT imaging: A versatile diphosphine chelator for (99m)Tc radiolabelling of peptides. Dalton Trans. 2021, 50, 16156–16165. [Google Scholar] [CrossRef]
  89. Kikuchi, T.; Okada, M.; Nengaki, N.; Furutsuka, K.; Wakizaka, H.; Okamura, T.; Zhang, M.R.; Kato, K. Efficient synthesis and chiral separation of 11C-labeled ibuprofen assisted by DMSO for imaging of in vivo behavior of the individual isomers by positron emission tomography. Bioorg. Med. Chem. 2011, 19, 3265–3273. [Google Scholar] [CrossRef]
  90. Nozaki, S.; Ozaki, N.; Suzuki, S.; Goto, M.; Mawatari, A.; Nakatani, Y.; Hayashinaka, E.; Wada, Y.; Doi, H.; Watanabe, Y. Development of Diagnostic Techniques for Early Rheumatoid Arthritis Using Positron Emission Tomography with [(11)C]PK11195 and [(11)C]Ketoprofen Tracers. Mol. Imaging Biol. 2017, 19, 746–753. [Google Scholar] [CrossRef]
  91. Shrestha, S.; Kim, M.J.; Eldridge, M.; Lehmann, M.L.; Frankland, M.; Liow, J.S.; Yu, Z.X.; Cortes-Salva, M.; Telu, S.; Henter, I.D.; et al. PET measurement of cyclooxygenase-2 using a novel radioligand: Upregulation in primate neuroinflammation and first-in-human study. J. Neuroinflamm. 2020, 17, 140. [Google Scholar] [CrossRef] [PubMed]
  92. Beckford-Vera, D.R.; Gonzalez-Junca, A.; Janneck, J.S.; Huynh, T.L.; Blecha, J.E.; Seo, Y.; Li, X.; VanBrocklin, H.F.; Franc, B.L. PET/CT Imaging of Human TNFalpha Using [(89)Zr]Certolizumab Pegol in a Transgenic Preclinical Model of Rheumatoid Arthritis. Mol. Imaging Biol. 2020, 22, 105–114. [Google Scholar] [CrossRef] [PubMed]
  93. Carron, P.; Lambert, B.; Van Praet, L.; De Vos, F.; Varkas, G.; Jans, L.; Elewaut, D.; Van den Bosch, F. Scintigraphic detection of TNF-driven inflammation by radiolabelled certolizumab pegol in patients with rheumatoid arthritis and spondyloarthritis. RMD Open 2016, 2, e000265. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Lambert, B.; Carron, P.; D’Asseler, Y.; Bacher, K.; Van den Bosch, F.; Elewaut, D.; Verbruggen, G.; Beyaert, R.; Dumolyn, C.; De Vos, F. (99m)Tc-labelled S-HYNIC certolizumab pegol in rheumatoid arthritis and spondyloarthritis patients: A biodistribution and dosimetry study. EJNMMI Res. 2016, 6, 88. [Google Scholar] [CrossRef] [Green Version]
  95. Alexandre, D.J.A.; Carmo, C.C.M.; Romeiro, L.D.; Gutfilen-Schlesinger, G.; Amarante, J.L., Jr.; de Souza, S.A.L.; Gutfilen, B. 99mTc-antitumor necrosis factor-alpha scintigraphy for the detection of inflammatory activity in rheumatoid arthritis. Nucl. Med. Commun. 2021, 42, 389–395. [Google Scholar] [CrossRef]
  96. Conti, F.; Malviya, G.; Ceccarelli, F.; Priori, R.; Iagnocco, A.; Valesini, G.; Signore, A. Role of scintigraphy with (9)(9)mTc-infliximab in predicting the response of intraarticular infliximab treatment in patients with refractory monoarthritis. Eur. J. Nucl. Med. Mol. Imaging 2012, 39, 1339–1347. [Google Scholar] [CrossRef] [Green Version]
  97. van der Laken, C.J.; Voskuyl, A.E.; Roos, J.C.; Stigter van Walsum, M.; de Groot, E.R.; Wolbink, G.; Dijkmans, B.A.; Aarden, L.A. Imaging and serum analysis of immune complex formation of radiolabelled infliximab and anti-infliximab in responders and non-responders to therapy for rheumatoid arthritis. Ann. Rheum. Dis. 2007, 66, 253–256. [Google Scholar] [CrossRef] [Green Version]
  98. Wythe, S.E.; DiCara, D.; Taher, T.E.; Finucane, C.M.; Jones, R.; Bombardieri, M.; Man, Y.K.; Nissim, A.; Mather, S.J.; Chernajovsky, Y.; et al. Targeted delivery of cytokine therapy to rheumatoid tissue by a synovial targeting peptide. Ann. Rheum. Dis. 2013, 72, 129–135. [Google Scholar] [CrossRef] [Green Version]
  99. Giatromanolaki, A.; Sivridis, E.; Maltezos, E.; Athanassou, N.; Papazoglou, D.; Gatter, K.C.; Harris, A.L.; Koukourakis, M.I. Upregulated hypoxia inducible factor-1alpha and -2alpha pathway in rheumatoid arthritis and osteoarthritis. Arthritis Res. Ther. 2003, 5, R193. [Google Scholar] [CrossRef] [Green Version]
  100. Hitchon, C.; Wong, K.; Ma, G.; Reed, J.; Lyttle, D.; El-Gabalawy, H. Hypoxia-induced production of stromal cell-derived factor 1 (CXCL12) and vascular endothelial growth factor by synovial fibroblasts. Arthritis Rheum. 2002, 46, 2587–2597. [Google Scholar] [CrossRef]
  101. Wang, C.H.; Yao, H.; Chen, L.N.; Jia, J.F.; Wang, L.; Dai, J.Y.; Zheng, Z.H.; Chen, Z.N.; Zhu, P. CD147 induces angiogenesis through a vascular endothelial growth factor and hypoxia-inducible transcription factor 1 alpha-mediated pathway in rheumatoid arthritis. Arthritis Rheum. 2012, 64, 1818–1827. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Westra, J.; Brouwer, E.; van Roosmalen, I.A.; Doornbos-van der Meer, B.; van Leeuwen, M.A.; Posthumus, M.D.; Kallenberg, C.G. Expression and regulation of HIF-1alpha in macrophages under inflammatory conditions; significant reduction of VEGF by CaMKII inhibitor. BMC Musculoskelet. Disord. 2010, 11, 61. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Westra, J.; Molema, G.; Kallenberg, C.G. Hypoxia-inducible factor-1 as regulator of angiogenesis in rheumatoid arthritis-therapeutic implications. Curr. Med. Chem. 2010, 17, 254–263. [Google Scholar] [CrossRef]
  104. Konisti, S.; Kiriakidis, S.; Paleolog, E.M. Hypoxia--a key regulator of angiogenesis and inflammation in rheumatoid arthritis. Nat. Rev. Rheumatol. 2012, 8, 153–162. [Google Scholar] [CrossRef]
  105. Liu, J.; Hajibeigi, A.; Ren, G.; Lin, M.; Siyambalapitiyage, W.; Liu, Z.; Simpson, E.; Parkey, R.W.; Sun, X.; Oz, O.K. Retention of the radiotracers 64Cu-ATSM and 64Cu-PTSM in human and murine tumors is influenced by MDR1 protein expression. J. Nucl. Med. 2009, 50, 1332–1339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Adams, R.L.; Adams, I.P.; Lindow, S.W.; Zhong, W.; Atkin, S.L. Somatostatin receptors 2 and 5 are preferentially expressed in proliferating endothelium. Br. J. Cancer 2005, 92, 1493–1498. [Google Scholar] [CrossRef] [Green Version]
  107. Casnici, C.; Lattuada, D.; Crotta, K.; Truzzi, M.C.; Corradini, C.; Ingegnoli, F.; Tonna, N.; Bianco, F.; Marelli, O. Anti-inflammatory Effect of Somatostatin Analogue Octreotide on Rheumatoid Arthritis Synoviocytes. Inflammation 2018, 41, 1648–1660. [Google Scholar] [CrossRef]
  108. Pacilio, M.; Lauri, C.; Prosperi, D.; Petitti, A.; Signore, A. New SPECT and PET Radiopharmaceuticals for Imaging Inflammatory Diseases: A Meta-analysis of the Last 10 Years. Semin. Nucl. Med. 2018, 48, 261–276. [Google Scholar] [CrossRef]
  109. Reubi, J.C.; Waser, B.; Markusse, H.M.; Krenning, E.P.; VanHagen, M.; Laissue, J.A. Vascular somatostatin receptors in synovium from patients with rheumatoid arthritis. Eur. J. Pharmacol. 1994, 271, 371–378. [Google Scholar] [CrossRef]
  110. Watson, J.C.; Balster, D.A.; Gebhardt, B.M.; O’Dorisio, T.M.; O’Dorisio, M.S.; Espenan, G.D.; Drouant, G.J.; Woltering, E.A. Growing vascular endothelial cells express somatostatin subtype 2 receptors. Br. J. Cancer 2001, 85, 266–272. [Google Scholar] [CrossRef]
  111. Danesi, R.; Agen, C.; Benelli, U.; Paolo, A.D.; Nardini, D.; Bocci, G.; Basolo, F.; Campagni, A.; Tacca, M.D. Inhibition of experimental angiogenesis by the somatostatin analogue octreotide acetate (SMS 201–995). Clin. Cancer Res. 1997, 3, 265–272. [Google Scholar] [PubMed]
  112. Dasgupta, P. Somatostatin analogues: Multiple roles in cellular proliferation, neoplasia, and angiogenesis. Pharmacol. Therapeut. 2004, 102, 61–85. [Google Scholar] [CrossRef]
  113. Takeba, Y.; Suzuki, N.; Takeno, M.; Asai, T.; Tsuboi, S.; Hoshino, T.; Sakane, T. Modulation of synovial cell function by somatostatin in patients with rheumatoid arthritis. Arthritis Rheum. 1997, 40, 2128–2138. [Google Scholar] [CrossRef]
  114. Anzola, L.K.; Glaudemans, A.; Dierckx, R.; Martinez, F.A.; Moreno, S.; Signore, A. Somatostatin receptor imaging by SPECT and PET in patients with chronic inflammatory disorders: A systematic review. Eur. J. Nucl. Med. Mol. Imaging 2019, 46, 2496–2513. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Cui, N.; Hu, M.; Khalil, R.A. Biochemical and Biological Attributes of Matrix Metalloproteinases. Prog. Mol. Biol. Transl. Sci. 2017, 147, 1–73. [Google Scholar] [PubMed] [Green Version]
  116. Cabral-Pacheco, G.A.; Garza-Veloz, I.; Castruita-De la Rosa, C.; Ramirez-Acuna, J.M.; Perez-Romero, B.A.; Guerrero-Rodriguez, J.F.; Martinez-Avila, N.; Martinez-Fierro, M.L. The Roles of Matrix Metalloproteinases and Their Inhibitors in Human Diseases. Int. J. Mol. Sci. 2020, 21, 9739. [Google Scholar] [CrossRef] [PubMed]
  117. Sun, C.; Feng, S.B.; Cao, Z.W.; Bei, J.J.; Chen, Q.; Zhao, W.B.; Xu, X.J.; Zhou, Z.; Yu, Z.P.; Hu, H.Y. Up-Regulated Expression of Matrix Metalloproteinases in Endothelial Cells Mediates Platelet Microvesicle-Induced Angiogenesis. Cell Physiol. Biochem. 2017, 41, 2319–2332. [Google Scholar] [CrossRef]
  118. Middleton, J.; Americh, L.; Gayon, R.; Julien, D.; Aguilar, L.; Amalric, F.; Girard, J.P. Endothelial cell phenotypes in the rheumatoid synovium: Activated, angiogenic, apoptotic and leaky. Arthritis Res. Ther. 2004, 6, 60–72. [Google Scholar] [CrossRef] [Green Version]
  119. Jungel, A.; Ospelt, C.; Lesch, M.; Thiel, M.; Sunyer, T.; Schorr, O.; Michel, B.A.; Gay, R.E.; Kolling, C.; Flory, C.; et al. Effect of the oral application of a highly selective MMP-13 inhibitor in three different animal models of rheumatoid arthritis. Ann. Rheum. Dis. 2010, 69, 898–902. [Google Scholar] [CrossRef] [Green Version]
  120. Rundhaug, J.E. Matrix metalloproteinases, angiogenesis, and cancer: Commentary re: A. C. Lockhart et al., Reduction of wound angiogenesis in patients treated with BMS-275291, a broad spectrum matrix metalloproteinase inhibitor. Clin. Cancer Res. 2003, 9, 551–554. [Google Scholar]
  121. Zhou, M.; Qin, S.; Chu, Y.; Wang, F.; Chen, L.; Lu, Y. Immunolocalization of MMP-2 and MMP-9 in human rheumatoid synovium. Int. J. Clin. Exp. Pathol. 2014, 7, 3048–3056. [Google Scholar] [PubMed]
  122. Jackson, C.; Nguyen, M.; Arkell, J.; Sambrook, P. Selective matrix metalloproteinase (MMP) inhibition in rheumatoid arthritis--targetting gelatinase A activation. Inflamm. Res. 2001, 50, 183–186. [Google Scholar] [CrossRef] [PubMed]
  123. Kim, K.S.; Choi, H.M.; Lee, Y.A.; Choi, I.A.; Lee, S.H.; Hong, S.J.; Yang, H.I.; Yoo, M.C. Expression levels and association of gelatinases MMP-2 and MMP-9 and collagenases MMP-1 and MMP-13 with VEGF in synovial fluid of patients with arthritis. Rheumatol. Int. 2011, 31, 543–547. [Google Scholar] [CrossRef]
  124. Kim, K.S.; Lee, Y.A.; Choi, H.M.; Yoo, M.C.; Yang, H.I. Implication of MMP-9 and urokinase plasminogen activator (uPA) in the activation of pro-matrix metalloproteinase (MMP)-13. Rheumatol. Int. 2012, 32, 3069–3075. [Google Scholar] [CrossRef]
  125. Itoh, T.; Matsuda, H.; Tanioka, M.; Kuwabara, K.; Itohara, S.; Suzuki, R. The role of matrix metalloproteinase-2 and matrix metalloproteinase-9 in antibody-induced arthritis. J. Immunol. 2002, 169, 2643–2647. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  126. Mengshol, J.A.; Mix, K.S.; Brinckerhoff, C.E. Matrix metalloproteinases as therapeutic targets in arthritic diseases-Bull’s-eye or missing the mark? Arthritis Rheum. 2002, 46, 13–20. [Google Scholar] [CrossRef]
  127. Breyholz, H.J.; Schafers, M.; Wagner, S.; Holtke, C.; Faust, A.; Rabeneck, H.; Levkau, B.; Schober, O.; Kopka, K. C-5-disubstituted barbiturates as potential molecular probes for noninvasive matrix metalloproteinase imaging. J. Med. Chem. 2005, 48, 3400–3409. [Google Scholar] [CrossRef]
  128. Breyholz, H.J.; Wagner, S.; Faust, A.; Riemann, B.; Holtke, C.; Hermann, S.; Schober, O.; Schafers, M.; Kopka, K. Radiofluorinated pyrimidine-2,4,6-triones as molecular probes for noninvasive MMP-targeted imaging. ChemMedChem 2010, 5, 777–789. [Google Scholar] [CrossRef]
  129. Eming, S.A.; Hubbell, J.A. Extracellular matrix in angiogenesis: Dynamic structures with translational potential. Exp. Dermatol 2011, 20, 605–613. [Google Scholar] [CrossRef]
  130. Bencharit, S.; Bin Cui, C.; Siddiqui, A.; Howard-Williams, E.L.; Sondek, J.; Zuobi-Hasona, K.; Aukhil, I. Structural insights into fibronectin type III domain-mediated signaling. J. Mol. Biol. 2007, 367, 303–309. [Google Scholar] [CrossRef] [Green Version]
  131. Kumra, H.; Reinhardt, D.P. Fibronectin-targeted drug delivery in cancer. Adv. Drug Deliv. Rev. 2016, 97, 101–110. [Google Scholar] [CrossRef] [PubMed]
  132. Kriegsmann, J.; Berndt, A.; Hansen, T.; Borsi, L.; Zardi, L.; Brauer, R.; Petrow, P.K.; Otto, M.; Kirkpatrick, C.J.; Gay, S.; et al. Expression of fibronectin splice variants and oncofetal glycosylated fibronectin in the synovial membranes of patients with rheumatoid arthritis and osteoarthritis. Rheumatol. Int. 2004, 24, 25–33. [Google Scholar] [CrossRef] [PubMed]
  133. Di Matteo, A.; Belloni, E.; Pradella, D.; Cappelletto, A.; Volf, N.; Zacchigna, S.; Ghigna, C. Alternative splicing in endothelial cells: Novel therapeutic opportunities in cancer angiogenesis. J. Exp. Clin. Cancer Res. 2020, 39, 275. [Google Scholar] [CrossRef] [PubMed]
  134. Schwager, K.; Kaspar, M.; Bootz, F.; Marcolongo, R.; Paresce, E.; Neri, D.; Trachsel, E. Preclinical characterization of DEKAVIL (F8-IL10), a novel clinical-stage immunocytokine which inhibits the progression of collagen-induced arthritis. Arthritis Res. Ther. 2009, 11, R142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  135. Hemmerle, T.; Doll, F.; Neri, D. Antibody-based delivery of IL4 to the neovasculature cures mice with arthritis. Proc. Natl. Acad. Sci. USA 2014, 111, 12008–12012. [Google Scholar] [CrossRef] [Green Version]
  136. Berndt, A.; Borsi, L.; Luo, X.; Zardi, L.; Katenkamp, D.; Kosmehl, H. Evidence of ED-B+ fibronectin synthesis in human tissues by non-radioactive RNA in situ hybridization. Investigations on carcinoma (oral squamous cell and breast carcinoma), chronic inflammation (rheumatoid synovitis) and fibromatosis (Morbus Dupuytren). Histochem. Cell Biol. 1998, 109, 249–255. [Google Scholar] [CrossRef]
  137. Vollmer, S.; Vater, A.; Licha, K.; Gemeinhardt, I.; Gemeinhardt, O.; Voigt, J.; Ebert, B.; Schnorr, J.; Taupitz, M.; Macdonald, R.; et al. Extra domain B fibronectin as a target for near-infrared fluorescence imaging of rheumatoid arthritis affected joints in vivo. Mol. Imaging 2009, 8, 330–340. [Google Scholar] [CrossRef] [Green Version]
  138. Galeazzi, M.; Bazzichi, L.; Sebastiani, G.D.; Neri, D.; Garcia, E.; Ravenni, N.; Giovannoni, L.; Wilton, J.; Bardelli, M.; Baldi, C.; et al. A phase IB clinical trial with Dekavil (F8-IL10), an immunoregulatory ’armed antibody’ for the treatment of rheumatoid arthritis, used in combination wiIh methotrexate. Isr. Med. Assoc. J. 2014, 16, 666. [Google Scholar]
  139. Harjunpaa, H.; Llort Asens, M.; Guenther, C.; Fagerholm, S.C. Cell Adhesion Molecules and Their Roles and Regulation in the Immune and Tumor Microenvironment. Front. Immunol. 2019, 10, 1078. [Google Scholar] [CrossRef] [Green Version]
  140. Bischoff, J. Cell adhesion and angiogenesis. J. Clin. Investig. 1997, 99, 373–376. [Google Scholar] [CrossRef] [Green Version]
  141. Malemud, C.J.; Reddy, S.K. Targeting Cytokines, Chemokines and Adhesion Molecules in Rheumatoid Arthritis. Curr. Rheumatol. Rev. 2008, 4, 219–234. [Google Scholar] [CrossRef]
  142. Jalkanen, S.; Karikoski, M.; Mercier, N.; Koskinen, K.; Henttinen, T.; Elima, K.; Salmivirta, K.; Salmi, M. The oxidase activity of vascular adhesion protein-1 (VAP-1) induces endothelial E- and P-selectins and leukocyte binding. Blood 2007, 110, 1864–1870. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  143. Eliceiri, B.P.; Cheresh, D.A. The role of alphav integrins during angiogenesis. Mol. Med. 1998, 4, 741–750. [Google Scholar] [CrossRef] [Green Version]
  144. Francavilla, C.; Maddaluno, L.; Cavallaro, U. The functional role of cell adhesion molecules in tumor angiogenesis. Semin. Cancer Biol. 2009, 19, 298–309. [Google Scholar] [CrossRef] [PubMed]
  145. Aalto, K.; Autio, A.; Kiss, E.A.; Elima, K.; Nymalm, Y.; Veres, T.Z.; Marttila-Ichihara, F.; Elovaara, H.; Saanijoki, T.; Crocker, P.R.; et al. Siglec-9 is a novel leukocyte ligand for vascular adhesion protein-1 and can be used in PET imaging of inflammation and cancer. Blood 2011, 118, 3725–3733. [Google Scholar] [CrossRef]
  146. Salmi, M.; Jalkanen, S. Vascular Adhesion Protein-1: A Cell Surface Amine Oxidase in Translation. Antioxid. Redox. 2019, 30, 314–332. [Google Scholar] [CrossRef] [PubMed]
  147. Marttila-Ichihara, F.; Smith, D.J.; Stolen, C.; Yegutkin, G.G.; Elima, K.; Mercier, N.; Kiviranta, R.; Pihlavisto, M.; Alaranta, S.; Pentikainen, U.; et al. Vascular amine oxidases are needed for leukocyte extravasation into inflamed joints in vivo. Arthritis Rheum. 2006, 54, 2852–2862. [Google Scholar] [CrossRef] [PubMed]
  148. Horvath, A.; Menghis, A.; Botz, B.; Borbely, E.; Kemeny, A.; Tekus, V.; Csepregi, J.Z.; Mocsai, A.; Juhasz, T.; Zakany, R.; et al. Analgesic and Anti-Inflammatory Effects of the Novel Semicarbazide-Sensitive Amine-Oxidase Inhibitor SzV-1287 in Chronic Arthritis Models of the Mouse. Sci. Rep. 2017, 7, 39863. [Google Scholar] [CrossRef]
  149. Bournazou, E.; Samuels, J.; Zhou, H.; Krasnokutsky, S.; Patel, J.; Han, T.Z.; Bencardino, J.; Rybak, L.; Abramson, S.B.; Junker, U.; et al. Vascular Adhesion Protein-1 (VAP-1) as Predictor of Radiographic Severity in Symptomatic Knee Osteoarthritis in the New York University Cohort. Int. J. Mol. Sci. 2019, 20, 2642. [Google Scholar] [CrossRef] [Green Version]
  150. Tabi, T.; Szoko, E.; Merey, A.; Toth, V.; Matyus, P.; Gyires, K. Study on SSAO enzyme activity and anti-inflammatory effect of SSAO inhibitors in animal model of inflammation. Neural Transm. 2013, 120, 963–967. [Google Scholar] [CrossRef]
  151. Davis, L.S.; Sackler, M.; Brezinschek, R.I.; Lightfoot, E.; Bailey, J.L.; Oppenheimer-Marks, N.; Lipsky, P.E. Inflammation, immune reactivity, and angiogenesis in a severe combined immunodeficiency model of rheumatoid arthritis. Am. J. Pathol. 2002, 160, 357–367. [Google Scholar] [CrossRef] [Green Version]
  152. Li, H.; Du, S.; Niu, P.; Gu, X.; Wang, J.; Zhao, Y. Vascular Adhesion Protein-1 (VAP-1)/Semicarbazide-Sensitive Amine Oxidase (SSAO): A Potential Therapeutic Target for Atherosclerotic Cardiovascular Diseases. Front. Pharmacol. 2021, 12, 679707. [Google Scholar] [CrossRef] [PubMed]
  153. Salmi, M.; Jalkanen, S. Ectoenzymes in leukocyte migration and their therapeutic potential. Semin. Immunopathol. 2014, 36, 163–176. [Google Scholar] [CrossRef] [PubMed]
  154. Kivi, E.; Elima, K.; Aalto, K.; Nymalm, Y.; Auvinen, K.; Koivunen, E.; Otto, D.M.; Crocker, P.R.; Salminen, T.A.; Salmi, M.; et al. Human Siglec-10 can bind to vascular adhesion protein-1 and serves as its substrate. Blood 2009, 114, 5385–5392. [Google Scholar] [CrossRef]
  155. Ley, K. The role of selectins in inflammation and disease. Trends Mol. Med. 2003, 9, 263–268. [Google Scholar] [CrossRef] [Green Version]
  156. Koch, A.E.; Halloran, M.M.; Haskell, C.J.; Shah, M.R.; Polverini, P.J. Angiogenesis mediated by soluble forms of E-selectin and vascular cell adhesion molecule-1. Nature 1995, 376, 517–519. [Google Scholar] [CrossRef]
  157. Firestein, G.S. Starving the synovium: Angiogenesis and inflammation in rheumatoid arthritis. J. Clin. Investig. 1999, 103, 3–4. [Google Scholar] [CrossRef] [Green Version]
  158. Sakurai, D.; Tsuchiya, N.; Yamaguchi, A.; Okaji, Y.; Tsuno, N.H.; Kobata, T.; Takahashi, K.; Tokunaga, K. Crucial role of inhibitor of DNA binding/differentiation in the vascular endothelial growth factor-induced activation and angiogenic processes of human endothelial cells. J. Immunol. 2004, 173, 5801–5809. [Google Scholar] [CrossRef] [Green Version]
  159. Sharaki, O.A.; El-Guiziry, D.A.; Abou-Zeid, A.A.; El-Noueam, K.I.; Helal, A.E.; Gaballah, A.E. Clinical usefulness of basic fibroblast growth factor and E-selectin in patients with rheumatoid arthritis. Egypt. Immunol. 2004, 11, 91–100. [Google Scholar]
  160. Chapman, P.T.; Jamar, F.; Keelan, E.T.; Peters, A.M.; Haskard, D.O. Use of a radiolabeled monoclonal antibody against E-selectin for imaging of endothelial activation in rheumatoid arthritis. Arthritis Rheum. 1996, 39, 1371–1375. [Google Scholar] [CrossRef]
  161. Morshed, A.; Abbas, A.B.; Hu, J.; Xu, H. Shedding New Light on The Role of alphanubeta3 and alpha5beta1 Integrins in Rheumatoid Arthritis. Molecules 2019, 24, 1537. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Kapp, T.G.; Rechenmacher, F.; Neubauer, S.; Maltsev, O.V.; Cavalcanti-Adam, E.A.; Zarka, R.; Reuning, U.; Notni, J.; Wester, H.J.; Mas-Moruno, C.; et al. Comprehensive Evaluation of the Activity and Selectivity Profile of Ligands for RGD-binding Integrins. Sci. Rep. 2017, 7, 39805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Lowin, T.; Straub, R.H. Integrins and their ligands in rheumatoid arthritis. Arthritis Res. Ther. 2011, 13, 244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Shahrara, S.; Castro-Rueda, H.P.; Haines, G.K.; Koch, A.E. Differential expression of the FAK family kinases in rheumatoid arthritis and osteoarthritis synovial tissues. Arthritis Res. Ther. 2007, 9, R112. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Wilder, R.L. Integrin alpha V beta 3 as a target for treatment of rheumatoid arthritis and related rheumatic diseases. Ann. Rheum. Dis. 2002, 61 (Suppl. S2), ii96–ii99. [Google Scholar] [CrossRef]
  166. Hao, J.; Wu, X.; Setrerrahmane, S.; Qian, K.; Hou, Y.; Yu, L.; Lin, C.; Wu, Q.; Xu, H. Combination Therapy of PEG-HM-3 and Methotrexate Retards Adjuvant-Induced Arthritis. Int J. Mol. Sci. 2017, 18, 1538. [Google Scholar] [CrossRef] [Green Version]
  167. Liu, Y.; Pan, Y.F.; Xue, Y.Q.; Fang, L.K.; Guo, X.H.; Guo, X.; Liu, M.; Mo, B.Y.; Yang, M.R.; Liu, F.; et al. uPAR promotes tumor-like biologic behaviors of fibroblast-like synoviocytes through PI3K/Akt signaling pathway in patients with rheumatoid arthritis. Cell Mol. Immunol. 2018, 15, 171–181. [Google Scholar] [CrossRef]
  168. Connolly, M.; Veale, D.J.; Fearon, U. Acute serum amyloid A regulates cytoskeletal rearrangement, cell matrix interactions and promotes cell migration in rheumatoid arthritis. Ann. Rheum. Dis. 2011, 70, 1296–1303. [Google Scholar] [CrossRef]
  169. Park, C.C.; Morel, J.C.M.; Amin, M.A.; Connors, M.A.; Harlow, L.A.; Koch, A.E. Evidence of IL-18 as a novel angiogenic mediator. J. Immunol. 2001, 167, 1644–1653. [Google Scholar] [CrossRef] [Green Version]
  170. Colman, R.W. Regulation of angiogenesis by the kallikrein-kinin system. Curr. Pharm. Des. 2006, 12, 2599–2607. [Google Scholar] [CrossRef]
  171. Chen, H.; Niu, G.; Wu, H.; Chen, X. Clinical Application of Radiolabeled RGD Peptides for PET Imaging of Integrin alphavbeta3. Theranostics 2016, 6, 78–92. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Notni, J.; Steiger, K.; Hoffmann, F.; Reich, D.; Kapp, T.G.; Rechenmacher, F.; Neubauer, S.; Kessler, H.; Wester, H.J. Complementary, Selective PET Imaging of Integrin Subtypes alpha5beta1 and alphavbeta3 Using 68Ga-Aquibeprin and 68Ga-Avebetrin. J. Nucl. Med. 2016, 57, 460–466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Claria, J.; Romano, M. Pharmacological intervention of cyclooxygenase-2 and 5-lipoxygenase pathways. Impact on inflammation and cancer. Curr. Pharm. Des. 2005, 11, 3431–3447. [Google Scholar] [CrossRef] [PubMed]
  174. Ma, Y.; Hong, F.F.; Yang, S.L. Role of prostaglandins in rheumatoid arthritis. Clin. Exp. Rheumatol. 2021, 39, 162–172. [Google Scholar] [CrossRef]
  175. Martel-Pelletier, J.; Pelletier, J.P.; Fahmi, H. Cyclooxygenase-2 and prostaglandins in articular tissues. Semin. Arthritis Rheum. 2003, 33, 155–167. [Google Scholar] [CrossRef]
  176. Ben-Av, P.; Crofford, L.J.; Wilder, R.L.; Hla, T. Induction of vascular endothelial growth factor expression in synovial fibroblasts by prostaglandin E and interleukin-1: A potential mechanism for inflammatory angiogenesis. FEBS Lett. 1995, 372, 83–87. [Google Scholar] [CrossRef] [Green Version]
  177. Lainer, D.T.; Brahn, E. New antiangiogenic strategies for the treatment of proliferative synovitis. Expert Opin. Investig. Drugs 2005, 14, 1–17. [Google Scholar] [CrossRef]
  178. El-Sayed, R.M.; Moustafa, Y.M.; El-Azab, M.F. Evening primrose oil and celecoxib inhibited pathological angiogenesis, inflammation, and oxidative stress in adjuvant-induced arthritis: Novel role of angiopoietin-1. Inflammopharmacology 2014, 22, 305–317. [Google Scholar] [CrossRef]
  179. Fikry, E.M.; Gad, A.M.; Eid, A.H.; Arab, H.H. Caffeic acid and ellagic acid ameliorate adjuvant-induced arthritis in rats via targeting inflammatory signals, chitinase-3-like protein-1 and angiogenesis. Pharmacother 2019, 110, 878–886. [Google Scholar] [CrossRef]
  180. Pakneshan, P.; Birsner, A.E.; Adini, I.; Becker, C.M.; D’Amato, R.J. Differential suppression of vascular permeability and corneal angiogenesis by nonsteroidal anti-inflammatory drugs. Investig. Ophthalmol. Vis. Sci. 2008, 49, 3909–3913. [Google Scholar] [CrossRef] [Green Version]
  181. Ha, M.K.; Song, Y.H.; Jeong, S.J.; Lee, H.J.; Jung, J.H.; Kim, B.; Song, H.S.; Huh, J.E.; Kim, S.H. Emodin inhibits proinflammatory responses and inactivates histone deacetylase 1 in hypoxic rheumatoid synoviocytes. Biol. Pharm. Bull. 2011, 34, 1432–1437. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  182. Alam, J.; Jantan, I.; Bukhari, S.N.A. Rheumatoid arthritis: Recent advances on its etiology, role of cytokines and pharmacotherapy. Biomed. Pharmacother. 2017, 92, 615–633. [Google Scholar] [CrossRef] [PubMed]
  183. Garcia, S.; Krausz, S.; Ambarus, C.A.; Fernandez, B.M.; Hartkamp, L.M.; van Es, I.E.; Hamann, J.; Baeten, D.L.; Tak, P.P.; Reedquist, K.A. Tie2 signaling cooperates with TNF to promote the pro-inflammatory activation of human macrophages independently of macrophage functional phenotype. PLoS ONE 2014, 9, e82088. [Google Scholar] [CrossRef]
  184. DeBusk, L.M.; Chen, Y.; Nishishita, T.; Chen, J.; Thomas, J.W.; Lin, P.C. Tie2 receptor tyrosine kinase, a major mediator of tumor necrosis factor alpha-induced angiogenesis in rheumatoid arthritis. Arthritis Rheum. 2003, 48, 2461–2471. [Google Scholar] [CrossRef] [PubMed]
  185. Scott, B.B.; Zaratin, P.F.; Gilmartin, A.G.; Hansbury, M.J.; Colombo, A.; Belpasso, C.; Winkler, J.D.; Jackson, J.R. TNF-alpha modulates angiopoietin-1 expression in rheumatoid synovial fibroblasts via the NF-kappa B signalling pathway. Biochem. Biophys. Res. Commun. 2005, 328, 409–414. [Google Scholar] [CrossRef] [PubMed]
  186. Shibasaki, S.; Tsunemi, S.; Kitano, S.; Sekiguchi, M.; Sano, H.; Iwasaki, T. Differential regulation of c-Met signaling pathways for synovial cell function. Springerplus 2014, 3, 554. [Google Scholar] [CrossRef] [Green Version]
  187. Maini, R.N.; Taylor, P.C.; Paleolog, E.; Charles, P.; Ballara, S.; Brennan, F.M.; Feldmann, M. Anti-tumour necrosis factor specific antibody (infliximab) treatment provides insights into the pathophysiology of rheumatoid arthritis. Ann. Rheum. Dis. 1999, 58, I56–I60. [Google Scholar] [CrossRef]
  188. Feldmann, M.; Maini, R.N. Anti-TNF alpha therapy of rheumatoid arthritis: What have we learned? Annu. Rev. Immunol. 2001, 19, 163–196. [Google Scholar] [CrossRef]
  189. Kanakaraj, P.; Puffer, B.A.; Yao, X.T.; Kankanala, S.; Boyd, E.; Shah, R.R.; Wang, G.; Patel, D.; Krishnamurthy, R.; Kaithamana, S.; et al. Simultaneous targeting of TNF and Ang2 with a novel bispecific antibody enhances efficacy in an in vivo model of arthritis. MAbs 2012, 4, 600–613. [Google Scholar] [CrossRef] [Green Version]
  190. Lupia, E.; Montrucchio, G.; Battaglia, E.; Modena, V.; Camussi, G. Role of tumor necrosis factor-alpha and platelet-activating factor in neoangiogenesis induced by synovial fluids of patients with rheumatoid arthritis. Eur. J. Immunol. 1996, 26, 1690–1694. [Google Scholar] [CrossRef]
  191. Tak, P.P.; Taylor, P.C.; Breedveld, F.C.; Smeets, T.J.; Daha, M.R.; Kluin, P.M.; Meinders, A.E.; Maini, R.N. Decrease in cellularity and expression of adhesion molecules by anti-tumor necrosis factor alpha monoclonal antibody treatment in patients with rheumatoid arthritis. Arthritis Rheum. 1996, 39, 1077–1081. [Google Scholar] [CrossRef] [PubMed]
  192. Camussi, G.; Lupia, E. The future role of anti-tumour necrosis factor (TNF) products in the treatment of rheumatoid arthritis. Drugs 1998, 55, 613–620. [Google Scholar] [CrossRef] [PubMed]
  193. Sidibe, A.; Mannic, T.; Arboleas, M.; Subileau, M.; Gulino-Debrac, D.; Bouillet, L.; Jan, M.; Vandhuick, T.; Le Loet, X.; Vittecoq, O.; et al. Soluble VE-cadherin in rheumatoid arthritis patients correlates with disease activity: Evidence for tumor necrosis factor alpha-induced VE-cadherin cleavage. Arthritis Rheum. 2012, 64, 77–87. [Google Scholar] [CrossRef] [PubMed]
  194. Shu, Q.; Amin, M.A.; Ruth, J.H.; Campbell, P.L.; Koch, A.E. Suppression of endothelial cell activity by inhibition of TNFalpha. Arthritis Res. Ther. 2012, 14, R88. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  195. Kennedy, A.; Ng, C.T.; Chang, T.C.; Biniecka, M.; O’Sulliv’n, J.N.; Heffernan, E.; Fearon, U.; Veale, D.J. Tumor necrosis factor blocking therapy alters joint inflammation and hypoxia. Arthritis Rheum. 2011, 63, 923–932. [Google Scholar] [CrossRef]
  196. Kong, X.; Zhang, Y.; Liu, C.; Guo, W.; Li, X.; Su, X.; Wan, H.; Sun, Y.; Lin, N. Anti-angiogenic effect of triptolide in rheumatoid arthritis by targeting angiogenic cascade. PLoS ONE 2013, 8, e77513. [Google Scholar] [CrossRef]
  197. Liu, C.; Kong, X.; Li, X.; Guo, W.; Zhang, C.; Sun, Y.; Su, X.; Liu, X.; Lu, A.; Lin, N. Wen Luo Yin inhibits angiogenesis in collagen-induced arthritis rat model and in vitro. J. Ethnopharmacol. 2013, 149, 478–489. [Google Scholar] [CrossRef]
  198. Paleolog, E. Target effector role of vascular endothelium in the inflammatory response: Insights from the clinical trial of anti-TNF alpha antibody in rheumatoid arthritis. Mol. Pathol. 1997, 50, 225–233. [Google Scholar] [CrossRef] [Green Version]
  199. Huang, M.; Qiu, Q.; Zeng, S.; Xiao, Y.; Shi, M.; Zou, Y.; Ye, Y.; Liang, L.; Yang, X.; Xu, H. Niclosamide inhibits the inflammatory and angiogenic activation of human umbilical vein endothelial cells. Inflamm. Res. 2015, 64, 1023–1032. [Google Scholar] [CrossRef]
  200. Barrera, P.; Oyen, W.J.; Boerman, O.C.; van Riel, P.L. Scintigraphic detection of tumour necrosis factor in patients with rheumatoid arthritis. Ann. Rheum. Dis. 2003, 62, 825–828. [Google Scholar] [CrossRef] [Green Version]
  201. Chapman, A.P. PEGylated antibodies and antibody fragments for improved therapy: A review. Adv. Drug Deliv. Rev. 2002, 54, 531–545. [Google Scholar] [CrossRef]
  202. Zhang, T.; Ouyang, X.; Gou, S.; Zhang, Y.; Yan, N.; Chang, L.; Li, B.; Zhang, F.; Liu, H.; Ni, J. Novel synovial targeting peptide-sinomenine conjugates as a potential strategy for the treatment of rheumatoid arthritis. Int. J. Pharm. 2022, 617, 121628. [Google Scholar] [CrossRef] [PubMed]
  203. Lee, L.; Buckley, C.; Blades, M.C.; Panayi, G.; George, A.J.; Pitzalis, C. Identification of synovium-specific homing peptides by in vivo phage display selection. Arthritis Rheum. 2002, 46, 2109–2120. [Google Scholar] [CrossRef] [PubMed]
  204. Swidrowska-Jaros, J.; Smolewska, E. A fresh look at angiogenesis in juvenile idiopathic arthritis. Cent. Eur. J. Immunol. 2018, 43, 325–330. [Google Scholar] [CrossRef]
  205. Folkman, J. Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat. Med. 1995, 1, 27–31. [Google Scholar] [CrossRef]
  206. Rodriguez, E.; Boelaars, K.; Brown, K.; Eveline Li, R.J.; Kruijssen, L.; Bruijns, S.C.M.; van Ee, T.; Schetters, S.T.T.; Crommentuijn, M.H.W.; van der Horst, J.C.; et al. Sialic acids in pancreatic cancer cells drive tumour-associated macrophage differentiation via the Siglec receptors Siglec-7 and Siglec-9. Nat. Commun. 2021, 12, 1270. [Google Scholar] [CrossRef]
  207. Li, L.; Chen, X.; Yu, J.; Yuan, S. Preliminary Clinical Application of RGD-Containing Peptides as PET Radiotracers for Imaging Tumors. Front. Oncol. 2022, 12, 837952. [Google Scholar] [CrossRef]
  208. Steiger, K.; Quigley, N.G.; Groll, T.; Richter, F.; Zierke, M.A.; Beer, A.J.; Weichert, W.; Schwaiger, M.; Kossatz, S.; Notni, J. There is a world beyond alphavbeta3-integrin: Multimeric ligands for imaging of the integrin subtypes alphavbeta6, alphavbeta8, alphavbeta3, and alpha5beta1 by positron emission tomography. EJNMMI Res. 2021, 11, 106. [Google Scholar] [CrossRef]
  209. Bernhagen, D.; Jungbluth, V.; Gisbert Quilis, N.; Dostalek, J.; White, P.B.; Jalink, K.; Timmerman, P. High-Affinity alpha5beta1-Integrin-Selective Bicyclic RGD Peptides Identified via Screening of Designed Random Libraries. ACS Comb. Sci. 2019, 21, 598–607. [Google Scholar] [CrossRef]
  210. Lopes, S.; Ferreira, S.; Caetano, M. PET/CT in the Evaluation of Hypoxia for Radiotherapy Planning in Head and Neck Tumors: Systematic Literature Review. J. Nucl. Med. Technol. 2021, 49, 107–113. [Google Scholar] [CrossRef]
  211. Thijssen, V.L.; Postel, R.; Brandwijk, R.J.; Dings, R.P.; Nesmelova, I.; Satijn, S.; Verhofstad, N.; Nakabeppu, Y.; Baum, L.G.; Bakkers, J.; et al. Galectin-1 is essential in tumor angiogenesis and is a target for antiangiogenesis therapy. Proc. Natl. Acad. Sci. USA 2006, 103, 15975–15980. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  212. Wang, Y.; Song, L.; Sun, J.; Sui, Y.; Li, D.; Li, G.; Liu, J.; Shu, Q. Expression of Galectin-9 and correlation with disease activity and vascular endothelial growth factor in rheumatoid arthritis. Clin. Exp. Rheumatol. 2020, 38, 654–661. [Google Scholar] [PubMed]
  213. O’Brien, M.J.; Shu, Q.; Stinson, W.A.; Tsou, P.S.; Ruth, J.H.; Isozaki, T.; Campbell, P.L.; Ohara, R.A.; Koch, A.E.; Fox, D.A.; et al. A unique role for galectin-9 in angiogenesis and inflammatory arthritis. Arthritis Res. Ther. 2018, 20, 31. [Google Scholar] [CrossRef] [Green Version]
  214. Hara, A.; Niwa, M.; Noguchi, K.; Kanayama, T.; Niwa, A.; Matsuo, M.; Hatano, Y.; Tomita, H. Galectin-3 as a Next-Generation Biomarker for Detecting Early Stage of Various Diseases. Biomolecules 2020, 10, 389. [Google Scholar] [CrossRef] [Green Version]
  215. Lu, D.; Zhou, H.; Li, N.; Wang, Y.; Zhang, T.; Wang, F.; Liu, N.; Zhu, H.; Zhang, J.; Yang, Z.; et al. Galectin expression detected by (68)Ga-galectracer PET as a predictive biomarker of radiotherapy resistance. Eur. J. Nucl. Med. Mol. Imaging 2022, 49, 2746–2760. [Google Scholar] [CrossRef] [PubMed]
  216. Duet, M.; Liote, F. Somatostatin and somatostatin analog scintigraphy: Any benefits for rheumatology patients? Jt. Bone Spine 2004, 71, 530–535. [Google Scholar] [CrossRef]
  217. Ehlerding, E.B.; Sun, L.; Lan, X.; Zeng, D.; Cai, W. Dual-Targeted Molecular Imaging of Cancer. J. Nucl. Med. 2018, 59, 390–395. [Google Scholar] [CrossRef]
  218. Zheng, Y.; Wang, H.; Tan, H.; Cui, X.; Yao, S.; Zang, J.; Zhang, L.; Zhu, Z. Evaluation of Lung Cancer and Neuroendocrine Neoplasm in a Single Scan by Targeting Both Somatostatin Receptor and Integrin alphavbeta3. Clin. Nucl. Med. 2019, 44, 687–694. [Google Scholar] [CrossRef]
  219. Baart, V.M.; Houvast, R.D.; de Geus-Oei, L.F.; Quax, P.H.A.; Kuppen, P.J.K.; Vahrmeijer, A.L.; Sier, C.F.M. Molecular imaging of the urokinase plasminogen activator receptor: Opportunities beyond cancer. EJNMMI Res. 2020, 10, 87. [Google Scholar] [CrossRef]
  220. Huang, C.C.; Tseng, T.T.; Liu, S.C.; Lin, Y.Y.; Law, Y.Y.; Hu, S.L.; Wang, S.W.; Tsai, C.H.; Tang, C.H. S1P Increases VEGF Production in Osteoblasts and Facilitates Endothelial Progenitor Cell Angiogenesis by Inhibiting miR-16–5p Expression via the c-Src/FAK Signaling Pathway in Rheumatoid Arthritis. Cells 2021, 10, 2168. [Google Scholar] [CrossRef]
  221. Sun, M.; Deng, R.; Wang, Y.; Wu, H.; Zhang, Z.; Bu, Y.; Zhang, H. Sphingosine kinase 1/sphingosine 1-phosphate/sphingosine 1-phosphate receptor 1 pathway: A novel target of geniposide to inhibit angiogenesis. Life Sci. 2020, 256, 117988. [Google Scholar] [CrossRef] [PubMed]
  222. Prasad, V.P.; Wagner, S.; Keul, P.; Hermann, S.; Levkau, B.; Schafers, M.; Haufe, G. Synthesis of fluorinated analogues of sphingosine-1-phosphate antagonists as potential radiotracers for molecular imaging using positron emission tomography. Bioorgan. Med. Chem. 2014, 22, 5168–5181. [Google Scholar] [CrossRef] [PubMed]
  223. Liu, H.; Jin, H.J.; Yue, X.Y.; Han, J.B.; Baum, P.; Abendschein, D.R.; Tu, Z.D. PET Study of Sphingosine-1-Phosphate Receptor 1 Expression in Response to Vascular Inflammation in a Rat Model of Carotid Injury. Mol. Imaging 2017, 16, 1536012116689770. [Google Scholar] [CrossRef] [PubMed]
  224. Ye, M.; Gai, Y.; Ji, H.; Jiang, Y.; Qiao, P.; Wang, W.; Zhang, Y.; Xia, X.; Lan, X. A Novel Radioimmune (99m)Tc-Labeled Tracer for Imaging Sphingosine 1-Phosphate Receptor 1 in Tumor Xenografts: An In Vitro and In Vivo Study. Front. Immunol. 2021, 12, 660842. [Google Scholar] [CrossRef] [PubMed]
  225. Shaikh, R.S.; Schilson, S.S.; Wagner, S.; Hermann, S.; Keul, P.; Levkau, B.; Schafers, M.; Haufe, G. Synthesis and evaluation of fluorinated fingolimod (FTY720) analogues for sphingosine-1-phosphate receptor molecular imaging by positron emission tomography. J. Med. Chem. 2015, 58, 3471–3484. [Google Scholar] [CrossRef]
Figure 1. (A) Schematic view of an inflamed arthritic knee. (B) Detailed viewed of the inflamed RA synovium, particularly highlighting processes involved in angiogenesis.
Figure 1. (A) Schematic view of an inflamed arthritic knee. (B) Detailed viewed of the inflamed RA synovium, particularly highlighting processes involved in angiogenesis.
Ijms 23 07071 g001
Figure 2. (A) The general structural components of a radiotracer. (B) Ideal radiotracer properties.
Figure 2. (A) The general structural components of a radiotracer. (B) Ideal radiotracer properties.
Ijms 23 07071 g002
Figure 3. The flow diagram of the search and selection procedure of studies. (A) Overview of the articles found during the search for RA-angiogenesis targets. (B) Overview of the articles found during the search for nuclear imaging tracers in RA.
Figure 3. The flow diagram of the search and selection procedure of studies. (A) Overview of the articles found during the search for RA-angiogenesis targets. (B) Overview of the articles found during the search for nuclear imaging tracers in RA.
Ijms 23 07071 g003
Table 1. Overview of all of the investigated PET/SPECT and scintigraphy tracers targeting angiogenesis in RA.
Table 1. Overview of all of the investigated PET/SPECT and scintigraphy tracers targeting angiogenesis in RA.
TypeTargetRadiotracer NameImaging ModeDevelopmental PhaseYearRef.
Environmental
factor
 
Hypoxia
 
 
18F-FMISOPETAnimal2017/2019[60,61]
18F-FAZAPETAnimal2017[61]
67Cu-ATSMPETAnimal2021[62]
α-[123I]-IAZA
α-[123I]-IAZP
ScintigraphyHuman2002/2001[63]
[64]
SomatostatinSST268Ga- DOTANOCPETHuman2022[65]
99mTc-HYNIC-TOCScintigraphyHuman2016[66]
111In-DTPA-D-Phe-octreotide
(pentetreotide)
ScintigraphyHuman1994[67]
99mTc-EDDA/HYNIC-TOCScintigraphyHuman2012[66]
99mTc-depreotideScintigraphyHuman2017[68]
Matrix ProteasesMMP-2/-918F-2,4,6-trione-30PETAnimal2012[69]
MMP-1311C-MMP-13 inhibitor (2d)PETAnimal *2017[70,71]
68Ga-MMP13 inhibitor (2i)PETAnimal *2017[70,71]
18F-MMP-13 inhibitor (2a)PETAnimal *2017[70,71]
ECMED-A fibronectin124I-F8-IL10PETAnimal/Human2019[72]
Adhesion moleculesVAP-168Ga-DOTA-Siglec-9PETAnimal/Human2021/2015[73,74]
124I-BTT-1023SPECTAnimal2013[75]
E-selectin111In-anti-E-selectinScintigraphyAnimal2009[76]
99mTc-1.2B6-FabScintigraphyHuman1997/2002[77,78]
Integrin a5B168Ga-AquibeprinPETAnimal2019[39]
Integrin avB368Ga-AvebretinPETAnimal2019[39]
68Ga-PRGD2PETHuman2014[79]
68Ga-HP(3)-RGD(3)PETAnimal2017[80]
18F-galacto-RGDPETAnimal2007[81]
68Ga-NODAGA-RGDykPETHuman2021[82]
99mTc-HYNIC-3PRGD2ScintigraphyAnimal2015[83]
99mTc-3PRGD2ScintigraphyAnimal/Human2017[84]
99mTc-maraciclatideScintigraphyHuman2020[85]
111In-DOTA-E-[c(RGDfK)]2SPECTAnimal2016[86]
99mTc-3P4-RGD2SPECTAnimal2013[87]
99mTcO2(DP-RGD)2SPECTAnimal2021[88]
EnzymesCOX1–311C-IbuprofenPETAnimal2011[89]
11C-KetoprofenPETAnimal2017[90]
COX-211C-MC1PETHuman2020[91]
CytokinesTNF-α89Zr-Certolizumab PegolPETAnimal2020[92]
99mTc- certolizumab pegolScintigraphyHuman2016[93]
99mTc-S-HYNIC-certolizumab pegolScintigraphyHuman2016[94]
99mTc-adalimumabScintigraphyHuman2021[95]
99mTc-InfliximabScintigraphyHuman2007/2012[96,97]
OtherIL-4- ECs of inflamed tissue125I-IL-4-SyETPSPECTAnimal2013[98]
* Not an arthritis animal model.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Khodadust, F.; Ezdoglian, A.; Steinz, M.M.; van Beijnum, J.R.; Zwezerijnen, G.J.C.; Jansen, G.; Tas, S.W.; van der Laken, C.J. Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis. Int. J. Mol. Sci. 2022, 23, 7071. https://doi.org/10.3390/ijms23137071

AMA Style

Khodadust F, Ezdoglian A, Steinz MM, van Beijnum JR, Zwezerijnen GJC, Jansen G, Tas SW, van der Laken CJ. Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis. International Journal of Molecular Sciences. 2022; 23(13):7071. https://doi.org/10.3390/ijms23137071

Chicago/Turabian Style

Khodadust, Fatemeh, Aiarpi Ezdoglian, Maarten M. Steinz, Judy R. van Beijnum, Gerben J. C. Zwezerijnen, Gerrit Jansen, Sander W. Tas, and Conny J. van der Laken. 2022. "Systematic Review: Targeted Molecular Imaging of Angiogenesis and Its Mediators in Rheumatoid Arthritis" International Journal of Molecular Sciences 23, no. 13: 7071. https://doi.org/10.3390/ijms23137071

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop