Abstract
Contrast-enhanced magnetic resonance imaging (MRI) is a commonly used diagnostic tool. Compared to the standard gadolinium-based contrast agents, ferumoxytol (Feraheme, AMAG Pharmaceuticals, Waltham, MA), used as an alternative contrast medium, is feasible in patients with impaired renal function. Other attractive imaging features of intravenous (IV) ferumoxytol include a prolonged blood pool phase and delayed intracellular uptake. With its unique pharmacological, metabolic and imaging properties, ferumoxytol may play a crucial role in future MR imaging of the central nervous system (CNS), various organs outside the CNS, and the cardiovascular system. Preclinical and clinical studies have demonstrated the overall safety and effectiveness of this novel contrast agent with rarely occurring anaphylactoid reactions. The purpose of this review is to describe the general and organ specific properties of ferumoxytol, as well as the advantages and potential pitfalls associated with its use in MRI. In order to more fully demonstrate the applications of ferumoxytol throughout the body, an imaging atlas was created and is available as supplementary material online.
Keywords: nephrotoxicity, chronic kidney disease
Introduction
Purpose of this review
Since the Food and Drug Administration (FDA) approved ferumoxytol (Feraheme, AMAG Pharmaceuticals, Waltham, MA) to treat iron deficiency anemia in adults with chronic kidney disease (CKD) in 2009, the off label use of this iron oxide nanoparticle compound by clinicians and researchers as a magnetic resonance imaging (MRI) contrast agent has grown rapidly. Ferumoxytol-enhanced imaging is feasible in patients with impaired renal function, a patient population in whom both gadolinium and iodinated contrast agents are contraindicated. Other attractive imaging features of intravenous (IV) ferumoxytol include a prolonged blood pool phase and delayed intracellular uptake. Furthermore, since iron is a naturally occurring element in the body, the administered iron enters the body’s natural iron metabolic pathways. Thus, the use of ferumoxytol is not currently associated with concerns regarding long-term deposition, as is the case with brain deposition of gadolinium-containing agents (1–4).
There are two aims of this review: first, to draw attention to a viable option of contrast enhanced cross sectional imaging in patients with renal failure; therefore Kidney International was chosen for publication. Second, to highlight the growing body of literature exploring a variety of clinical indications that suggest ferumoxytol may have utility not only as alternative to gadolinium-based contrast agents (GBCA), but also as a specialized MRI contrast agent with unique properties.
The number of clinical trials using ferumoxytol as a contrast agent is increasing. The co-authors are experienced in CNS, body or cardiovascular imaging with ferumoxytol. Based on the recommendations of the co-authors, we collected the relevant articles in the literature focusing on ferumoxytol over other ultrasmall superparamagnetic iron oxides (USPIOs) and highlighted the clinical applications rather than preclinical investigations.
This article describes ferumoxytol administration, dosing, and timing for imaging applications, followed by organ specific utilizations. We have created an atlas that more fully describes the application of ferumoxytol throughout the body and a table of relevant clinical publications using ferumoxytol which both can be found as an online supplement.
Ferumoxytol for iron replacement
Intravenous iron supplementation is important in patients with CKD owing to poor gastric absorption of oral iron. Insufficient absorption is related to the upregulation of hepcidin that occurs in CKD (5), which is exacerbated by excessive iron losses.
Though widely used, iron dextran, iron sucrose, and iron sodium gluconate have significant clinical limitations. Over the last decade, three new IV iron preparations have been developed that display tighter iron binding, allowing greater doses of iron to be given in a single administration. Along with ferric carboxymaltose and iron isomaltoside-1000, ferumoxytol has entered the therapeutic arena as an effective iron supplement (5). Ferumoxytol has favorable physicochemical characteristics, potentially reducing amounts of circulating free iron (6). Multiple studies have reported effective and safe treatment with ferumoxytol in iron deficiency (7–9).
Ferumoxytol for MR imaging
Initially, ferumoxytol was developed as an MRI contrast agent due to its effectiveness in shortening T1 and T2 relaxation times. Licensing the drug as a therapeutic iron supplement was likely a strategic decision, but this compound still holds great potential as an MRI contrast agent. Satisfactory contrast enhanced imaging can be performed with doses as low as 1 mg/kg and as high as 510 mg total dose. See Figure 1 for the potential imaging phases following IV administration (Figure 1).
Figure 1.
Phases of GBCA (A) and ferumoxytol (B) enhancement. Unlike GBCA, ferumoxytol has a long intravascular half-life that results in a long blood pool phase prior to detectable contrast extravasation. GBCA (gadolinium based contrast agents). Images provided courtesy of Dr. Neuwelt.
Arterial/venous dynamic phase
T2* based dynamic susceptibility contrast (DSC) perfusion imaging in the brain requires only 1 mg/kg ferumoxytol, and provides parametric maps in the brain similar to those achieved with a standard dose of gadolinium (10). The lack of early contrast extravasation with ferumoxytol is beneficial in cardiovascular and peripheral vascular examinations, wherein T1-weighted images acquired using a 4 mg/kg bolus injection clearly show intravascular enhancement as hyperintense structures. Although there is no evidence that these small bolus injections carry higher risk of adverse events, rapid injection of ferumoxytol is currently not recommended by the FDA (11).
Blood pool phase
One of the major advantages of nanoparticle imaging is the relatively long circulating time, with ferumoxytol displaying a plasma half-life of 14–21 h. Even in highly permeable tumors, high resolution imaging of the intravascular space can be achieved without visible background tissue enhancement. Steady-state blood volume mapping of the brain is a T2* based technique that requires 3–7 mg/kg ferumoxytol (12, 13), while the T1 based steady-state angiography of the peripheral vessels is performed using 3–4 mg/kg (14).
Delayed phase
In brain lesions, slow leakage of ferumoxytol through the disrupted blood-brain barrier (BBB) results in MRI signal changes peaking around 24 h after ferumoxytol administration (15). T1- weighted MRI shows signal increase similar to that seen with gadolinium. Signal decrease on T2/T2*- weighted images may represent high local iron concentration and/or intracellular uptake, which has many useful applications outside of the central nervous system (CNS). Indeed, intracellular uptake of ferumoxytol in abdominal organs, lymph nodes and vascular walls can be used to effectively delineate pathology in these areas (16, 17).
It is important to note that if a subsequent MRI is needed within 72 hours, ferumoxytol contrast enhancement may still be present in brain pathologies several days following administration. (15) In addition, decreased signal intensity in liver, spleen and bone marrow scans may persist for several months prior to returning to baseline.
Ferumoxytol metabolism and clearance
Following extravasation, ferumoxytol nanoparticles are taken up by cells of the mononuclear phagocyte system (MPS, previously known as reticuloendothelial system, RES), primarily in the liver, spleen and bone marrow. In the brain, macrophages or astrocytes contribute to ferumoxytol clearance (18, 19). Within these phagocytic cells, the nanoparticles are stored in secondary lysosomes (Figure 2) (19). The carboxymethyldextran coating is cleaved by dextranase and the cleaved coating is fully excreted by the kidneys and/or eliminated through feces. The iron core is incorporated into the body’s iron stores and used for cell metabolism and hemoglobin synthesis. Unless the patient has known hemosiderosis or hemochromatosis, the administered iron during MRI examination (maximum 510 mg) is safe, and no overloading occurs (20).
Figure 2.

Ferumoxytol traffics from systemic circulation to reactive lesions: immunocompetent rat xenograft model demonstrate sex travascular accumulation of ferumoxytol that correlated to the presence of activated macrophages.
A) MRI signal enhancement occurred in xenografted but not saline-injected hemispheres. Blue arrows show marked hypointense signal on T1 MR that correlates with histologic findings of intense immunostaining for ferumoxytol in macrophages using dextran-1. Yellow arrows show typical hyperintense enhancement on T1 MRI that correlates with diffuse milder staining for ferumoxytol in brain parenchyma and activated astrocytes using dextran-1. White arrowhead shows no signal change after saline injection in contralateral control hemisphere.
B) Immunostaining demonstrates that ferumoxytol traffics from the systemic circulation to reactive CNS lesions. Immunostains performed 48 h after H460 non-small cell lung carcinoma inoculation into caudate nucleus and 24 h after IV ferumoxytol administration. A representative inflammatory lesion was immunostained with dextran-1 (Dx1) for the ferumoxytol coating (red), CD163 for macrophages (brown), and GFAP for astrocytes (brown). All cell nuclei were counterstained with hematoxylin. T depicts live tumor cells. Arrowhead depicts reactive cells located outside of the main lesion. Scale bars, 100 μm.
C) Electron microscopy of a ferumoxytol laden macrophage (left) and two astrocyte processes (right). Left image: Iron oxide nanoparticles appear as electron-dense particles measuring ~30 nm in diameter located cytoplasmically and clustering around lysosomes (L). Right image: Two astrocyte processes (AP) showing dispersed, electron-dense ferumoxytol nanoparticles. N depicts neurons. Insets in both photos show a 27–33 nm diameter particle. Magnification 6800X, scale bar, 1 μm.
From McConnell et al, 2016 used with permission from Nanomedicine: Nanotechnology, Biology and Medicine.
Table 1 summarizes the physical, pharmacokinetic and imaging properties of ferumoxytol as compared to gadolinium.
Table 1.
Comparison between ferumoxytol and gadolinium
| Feature | Ferumoxytol | Gd-DTPA (Magnevist) |
|---|---|---|
| Basic element | Iron oxide | Gadolinium(III) |
| Molecular composition | Iron oxide coated with semisynthetic carbohydrate | Gadolinium chelated with diethylenetriamine pentaacetic acid |
| Relaxometric properties at 1.5 T. mM−1 s−1, 37°C in water | r1=15, r2=89 (107) | r1=3.3, r2=3.9 (108) |
| Elimination plasma half-life | 14 h (109) | 1.6 h |
| Relative size of the particle | Around 30 nm (109) | 0.357 nm |
| Permeability to intact BBB | Minimal | Minimal |
| Typical times to peak enhancement (in brain lesions) | 24 h (110) | 3.5–25 min (111) |
| Signal change on T1-weighted sequence | Increased signal (signal decrease at very high concentrations) | Increased signal |
| Signal change on T2-weighted sequence | Decreased signal | Usually no change |
| Signal change on T2*-weighted sequence | Decreased signal | Decreased signal if given as a bolus |
| Distribution | Dynamic phase, blood pool phase, delayed phase (extra and intracellular) | Dynamic phase, extracellular phase |
| Imaging dose | 1–7 mg/kg | 0.1 mmol/kg |
| Excretion | Stored with the body’s iron reserve and used in hemopoesis. Coating with renal and faecal excretion. | Renal |
| Boxed warning | Potential hypersensitivity | Potential NSF |
Ferumoxytol safety
On March 30th, 2015, the FDA revised the prescribing information of Feraheme to include the addition of a boxed warning, which highlighted potential fatal and serious hypersensitivity reactions (HSR) including anaphylaxis. The warning emphasized the importance of trained personnel, appropriate medications being readily available, and monitoring patients for at least 30 minutes post administration to properly screen for HSR. In the three pre-marketing clinical trials of Feraheme (21–23) including 1164 patients, the aggregate rate of anaphylaxis was 0.2% (14). The post-marketing trials had even better results; the largest trial with 8666 patients showed a serious adverse events (SAE) rate of 0.2% and an anaphylaxis rate of 0.02% (24). Recently, several studies described the diagnostic use of ferumoxytol in the MRI suite and no SAEs were reported (20, 25–34). The frequency of ferumoxytol-related adverse events (10–14.6%) and SAEs (0–1%) were comparable in all investigations, with rates similar to those seen with ionic iodinated contrast agents, and were ten times higher than gadolinium-related events (35). The initial administration rate of a 510 mg bolus over 17 s was lowered to a slow infusion of 510 mg diluted ferumoxytol over 15 minutes as the FDA recommendation suggests. By reducing the rate of administration the frequency of these events may also be reduced.
Ferumoxytol has also shown an excellent safety profile in pediatric patients (20). Typical doses of 1–5 mg/kg for imaging purposes are much lower than the therapeutic dose and do not have a significant effect on hemoglobin values (20).
Central nervous system MR imaging with ferumoxytol
Table 2 and Figure 3 indicate the most commonly used imaging sequences in the CNS.
Table 2.
Applications of ferumoxytol in CNS imaging
| MRI sequences | Use | Advantages | Disadvantages | |
|---|---|---|---|---|
| Dynamic phase | DSC perfusion | DDx based on lesion vascularity pre-operative grading prognosis assessment biopsy guidance therapy monitoring progression vs. pseudoprogression detection of recurrence/malignant transformation stroke / hemodynamic assessment |
low dose (1 mg/kg) needed no early contrast extravasation CBV, CBF, TTP, MTT, similar to CT/MR (gadolinium) perfusion |
low spatial resolution susceptibility artifacts, distortions overestimation of large vessels (blooming) bolus administration needed |
| Blood pool phase | T2*-weighted (GRE) | improved visualization of abnormal vessels | improved visualization of small abnormal vessels no bolus injection needed no post processing needed part of most routine CNS protocols |
susceptibility artifact motion sensitivity |
| SS-CBV mapping (using pre- and post-Fe T2*w GRE scans) | DDx: ↑rCBV: HGG, PCNSL, metastasis ↓rCBV: LGG, demyelinaXon, toxoplasmosis, abscess pre-operative grading prognosis assessment biopsy guidance therapy monitoring detection of recurrence/malignant transformation progression vs pseudoprogression |
high spatial resolution thin slices, good 3D reformat for neuronavigation no distortions full brain coverage possible no bolus injection needed not influenced by circulation parameters repeatable potentially quantitative |
susceptibility artifact motion sensitivity co-registration of the pre and post contrast images needed time consuming based on coverage (5–10 min) |
|
| Delayed phase | T1 enhancement (SE or MPRAGE) High SI |
lesion visualization assessment of BBB damage DDx: meningioma vs metastasis intraoperative lesion assessment post-Sx residuum assessment without additional CA injection differentiating intra- and extracellular iron (39) |
potential alternative to GBCA similar enhancement patterns as gadolinium in most pathologies prolonged enhancement (intra- and postoperative imaging without additional contrast agent administration) | additional scanning needed 24 h post ferumoxytol administration high concentration of ferumoxytol may result in T1 signal drop if enhancement persists it may mimic intracerebral blood |
| T2 enhancement (T2w TSE) Low SI |
assessment of BBB damage dg: meningioma vs metastasis intraoperative lesion assessment improved biopsy targeting differentiating intra- and extracellular iron |
prolonged enhancement may indicate intracellular uptake of ferumoxytol | additional scanning needed 24 h post ferumoxytol administration if enhancement persists it may mimic intracerebral blood may be difficult to differentiate between intracellular iron vs. high concentration interstitial iron |
|
DSC: dynamic susceptibility contrast. T2*w: T2*-weighted. GRE: gradient echo. SS-CBV mapping: steady-state cerebral blood volume mapping. Fe: ferumoxytol. GBCA: gadolinium-based contrast agents. SE: spin echo. MPRAGE: magnetization-prepared rapid gradient-echo. T2 TSE: T2 weighted turbo spin echo, DDx: differential diagnosis. Sx: surgery. BBB: blood brain barrier, CA: contrast agent, HGG: high grade glioma, LGG: low grade glioma, PCNSL: primary central nervous system lymphoma. CBV: cerebral blood volume. CBF: cerebral blood flow. TTP: time to peak. MTT: mean transit time. CNS: central nervous system. 3D: three-dimensional.
Figure 3.
Possible sequences with ferumoxytol in the CNS during the dynamic (A), blood pool (B, C), and delayed (D, E) phases after administration in a patient with newly diagnosed PCNSL. A: CBV map calculated from DSC perfusion with ferumoxytol shows mildly elevated rCBV within the neoplasm. B: Susceptibility weighted image shows curvilinear branching hypointensities compatible withs abnormal tumor vasculature. C: SS-CBV map with high resolution shows increased blood volume. D: Axial T1 MR demonstrates typical enhancement 24 hours after ferumoxytol injection. E: Axial T2 MR shows marked hypointensity within the tumor 24 hours after ferumoxytol administration. Images provided courtesy of Dr. Neuwelt.
Intracranial neoplasms
The use of ferumoxytol in MRI of primary brain tumors has been extensively studied. While lesion visualization with ferumoxytol is generally similar to GBCAs, differences in enhancement patterns may help in differential diagnosis. Perfusion MRI and steady-state blood volume mapping may improve tumor grading by identifying the most malignant area for surgical targeting and therapy monitoring.
Lesion visualization
Contrast enhanced MRI is routinely performed for the diagnosis of brain tumors, where enhancement is the marker of BBB breakdown. Since extravasation of the large molecules is slow, parenchymal enhancement is best seen in the delayed phase, 24 h post ferumoxytol injection (15). Contrast enhancement is found on various clinically used T1-weighted MRI sequences, improves border delineation, and allows assessment of lesion internal morphology (36, 37) (Figure 4). In primary malignant brain tumors, contrast enhancement with ferumoxytol is comparable with GBCA enhancement. No significant difference was found in the number of enhancing masses when Gd-MRI and Fe-MRI were compared in various primary brain tumors and in metastatic lesions (38). Differences in GBCA and ferumoxytol enhancement size and intensity may be present (37), and may reflect differences in pathology, contrast agent dose or timing of imaging. No difference was found in enhancement size with the two contrast agents in metastatic lesions (38) or in untreated glioma patients (36). Decreased signal on T2-weighted images in the delayed phase may indicate high local ferumoxytol concentration or retention in tumor-associated macrophages. Delayed T1 and T2 enhancement together may help differentiate extracellular iron (e.g. tumor necrosis) from intracellular iron (solid tumor) (39). T2*-weighted scans in the blood pool phase have a potential added value to standard of care by improving visualization of abnormal vasculature (40, 41).
Figure 4.
Axial brain MRI of 19-year-old man who had undergone previous radiation and chemotherapy for glioblastoma multiforme, with clinical suspicion for pseudoprogression. A: Spin-echo (SE) T1 MR obtained 24 h after ferumoxytol administration shows moderately intense enhancement. B: SE T1 gadoteridol-enhanced MR shows concordant enhancement size and intensity. C: Magnetization-prepared rapid gradient-echo (MPRAGE) MR performed 24 h after ferumoxytol administration shows moderate (though less homogeneous) enhancement intensity compared with SE image. D: Gadoteridol-enhanced MPRAGE shows more homogeneous and greater enhancement intensity than the ferumoxytol-enhanced MPRAGE scan. Administered ferumoxytol dose was 510 mg (Hamilton et. al. 2011). From Hamilton et al. 2011, used with permission from AJR.
Differential diagnosis
Dural-based enhancing masses may represent benign or malignant disease and there is no way to effectively distinguish between these pathologies solely with GBCA. Preliminary data suggests delayed T1 enhancement with ferumoxytol may help distinguish between meningioma and dural metastases when used in addition to GBCA. While all dural metastases enhanced strongly with ferumoxytol and GBCA, meningiomas showed poor to no enhancement with ferumoxytol (Figure 5) (42).
Figure 5.
Axial T1-weighted post contrast MRI images in two different patients with similar appearing frontal midline lesions. The mass (yellow arrows) enhances similarly with both Gd and Fe in a patient with melanoma metastases (A, B). A meningioma (yellow arrowheads) only enhances with Gd (C), not Fe (D) Of note, the glioblastoma in the same images (black arrows) enhances avidly with both agents (C, D). Images provided courtesy of Dr. Neuwelt.
Tumefactive demyelinating lesions (TDLs) are large lesions usually accompanied by mass effect and abnormal enhancement, mimicking brain tumor, posing a particular diagnostic dilemma in patients with or without established diagnosis of multiple sclerosis (MS) (43). Definitive diagnosis is given after surgery and histopathological confirmation. A noninvasive marker is highly desirable to assess this diagnostic dilemma. Ferumoxytol uptake in inflammatory lesions appear on delayed T1- and T2-weighted images (44), furthermore perfusion MRI may help to differentiate tumors and demyelinating lesions with relative cerebral blood volume (rCBV) being lower in TDLs.
Blood volume based lesion assessment
Measurement of the blood volume in tissues can carry important information about the level of vascularization. Small molecular weight contrast agents can only estimate blood volume using dynamic first pass technique. An intravascular contrast agent allows measuring the blood volume using the steady-state technique, by calculating signal differences between pre- and post-contrast (intravascular) images. This has the benefit of high spatial resolution, since no rapid acquisition is necessary.
Dynamic susceptibility contrast perfusion-derived or steady-state CBV maps reflect brain tumor malignancy by revealing hypervascular, highly perfused tumor regions. rCBV (relative to normal reference region) has been shown to correlate with survival and facilitates preoperative diagnosis by differentiating low and high grade tumors (45). Moreover, elevated rCBV values can predict transformation of low grade gliomas into high grade tumors 12 months before T1 enhancement appears (46). Although these studies were performed using GBCA, ferumoxytol-derived rCBV values were in agreement with GBCA-derived values (10).
High grade gliomas, metastases, and to a lesser degree primary CNS lymphomas, exhibit high rCBV values with DSC or steady-state imaging, which can help differentiate them from demyelination, abscesses, and toxoplasmosis.
Intraoperative MRI and surgical targeting
More aggressive, hypervascular tumor regions with high rCBV values are the optimal sites for biopsy and can be identified using dynamic or steady-state perfusion imaging with ferumoxytol. Dynamic perfusion methods, however, have limited ability to differentiate between vessels and high tissue CBV, which compromises the assessment of small active tumor hotspots and could be the source of subsequent tumor progression and short survival. Due to its near distortion free high resolution images, steady-state CBV mapping can overcome this limitation and may enable more precise targeting (Figure 6). Late T2 enhancement with ferumoxytol has also been described to mark specific tumor areas and may again be useful for accurate biopsy targeting (44). Finally, delayed enhancement with ferumoxytol enables intra-and post-operative assessment of residual tumor without injecting additional contrast agent.
Figure 6.
SS-CBV maps using ferumoxytol offer higher spatial resolution and allow better identification of hypervascular areas for surgical targeting in glioblastoma patients. Compare spatial resolution of cerebral blood volume (CBV) maps using DSC PWI with a standard dose of gadolinium (DSC-CBV) (left) and SS ferumoxytol CBV map using ferumoxytol (3 mg/kg). The scan on the right more clearly demonstrates a central hypervascular area in the right occipital hemisphere with the greatest vascularity, which is likely the most malignant portion of the tumor. Images provided courtesy of Dr. Neuwelt.
Treatment monitoring
After brain tumor therapy with radiation or chemoradiotherapy, increased edema and contrast enhancement on MRI may either represent tumor progression (growing tumor mass, indicating failure of ongoing therapy) or pseudoprogression, which is defined as a treatment-induced subacute inflammatory reaction without underlying tumor growth (47). Differentiating pseudoprogression from true tumor progression is a significant clinical problem. Response assessment according to the currently standard response assessment in neuro-oncology (RANO) criteria is based on morphology and contrast enhancement and may delay or prevent proper therapy (48). Similarly, antiangiogenic drugs affect BBB permeability without decreasing the tumor mass itself, which can lead to pseudoresponse (49) that cannot be differentiated from true response with the RANO criteria. It is becoming apparent that a phenomenon similar to pseudoprogression occurs after immune checkpoint blockade for cancer therapies (50). This has led to the development of immune-related response criteria to better identify cases of pseudoprogression (51), though these criteria are not specific to the CNS. According to the iRANO criteria published in 2015, patients who have imaging findings that meet RANO criteria for progressive disease within 6 months of starting immunotherapy should have a confirmation of follow up imaging (in 3 months) before defining the patients as non-responsive to treatment (52). The use of CBV mapping with ferumoxytol may help determine therapeutic efficacy in a variety of CNS tumors by differentiating highly vascular malignant tumor tissue from treatment related neuroinflammation, which correlates with survival (Figures 7 and 8) (10, 53).
Figure 7.
Pseudoprogression can be diagnosed using perfusion MRI with ferumoxytol and correlates with OS.
A) Axial MR images of a 47-year-old woman with glioblastoma. Patient underwent T1-weighted sequences post Gd and T2-weighted postoperative images prior to CRT. Eight days after CRT completion patient scans showed radiographic worsening followed by further deterioration on follow-up MRI while the patient continued to receive adjuvant temozolomide chemotherapy. Though updated Response Assessment in Neuro-Oncology Working Group (RANO) criteria would indicate true tumor progression, the blood volume of the lesion was low on ferumoxytol DSC-CBV maps, which instead indicates pseudoprogression. The patient received only 3 courses of bevacizumab and continued adjuvant temozolomide. Substantial improvement is seen on 5-month follow-up MRI after completion of bevacizumab therapy. Seven years after completion of CRT, the image indicates that the patient is stable without evidence of recurrence/progression (the patient is still on adjuvant temozolomide chemotherapy).
B) Kaplan-Meier estimates of overall survival by the presence or absence of pseudoprogression. CRT: Chemoradiotherapy.
From Gharamanov et al. 2014, modified with permission from CNS Oncology.
Figure 8.
42 year old male patient with glioblastoma. T1-weighted post gadoteridol scan shows no/minimal enhancement. In contrast, a highly vascular area (arrow) is seen on the high resolution steady-state CBV maps obtained with ferumoxytol. Residual tumor with high CBV shows reduction post chemoradiation therapy (post-surgery scan), and continued decrease on 1 month post chemoradiation scan, indicating treatment response.
Images provided courtesy of Dr. Neuwelt.
We can now obtain high-resolution steady-state-CBV images that differentiate regions of high vascularity and active tumor growth (10, 13). Steady-state blood volume mapping with ferumoxytol is particularly helpful in imaging cortical lesions with improved spatial resolution.
Neuroinflammation and demyelination
Delayed imaging with ferumoxytol allows the assessment of inflammation as an imaging biomarker. Few human MS investigations are performed using ferumoxytol, but studies using other ultrasmall paramagnetic iron oxides (USPIO), such as ferumoxtran-10, have identified MS lesion phenotypes with the following enhancement patterns: 1. GBCA+/delayed USPIO enhancement (USPIO+); 2. GBCA+/USPIO−; 3. GBCA−/USPIO+. The USPIO-specific lesions are of particular interest because they represent endothelial activation and diapedesis without BBB disruption. Longitudinal studies demonstrate that in a fraction of GBCA−/USPIO+ MS lesions, delayed USPIO preceded GBCA enhancement by one month, suggesting extensive monocyte/macrophage extravasation in MS (54).
Ferumoxytol has been used to study neuroinflammatory processes associated with Japanese macaque encephalomyelitis (JME) (55), a demyelinating disease of the non-human primate with strong similarities to MS (56). A study by Tagge et al. demonstrated substantially greater USPIO+ lesion volume than GBCA+ lesion volume during the acute JME disease phase, indicating spatially and temporally extensive monocyte/macrophage migration into CNS (55).
Farrell et al. demonstrated delayed enhancement with ferumoxytol in patients with MS, primary CNS lymphoma, posttransplant lymphoproliferative disorder, acute disseminated encephalomyelitis (ADEM) (Figure 9) (44) and chronic encephalitis; a higher number of lesions are visible with ferumoxytol, than with GBCAs.
Figure 9.
A) Patient with the suspicion of pontine glioma was diagnosed with pontine demyelination using ferumoxytol. Precontrast T1-weighted imaging shows little abnormality.
B) T1-weighted sagittal MRI 24-hours post-USPIO shows diffuse intense enhancement due to ferumoxytol uptake in the pons (arrow).
C) Axial noncontrast T2-weighted MR shows patchy nonspecific increased T2 signal within the pons (arrow).
D) Axial T2-weighted MR obtained 24 hours post-ferumoxytol shows patchy hypointensities in the pons (arrow) correlating with ferumoxytol uptake.
E) Ferumoxytol-based DSC perfusion imaging shows low rCBV centrally within the pons (arrow) corresponding to the area of enhancing abnormality that is similar to normal-appearing white matter, a finding supportive of demyelination rather than high grade primary CNS neoplasm. DSC = dynamic susceptibility-weighted contrast-enhanced; GBCA = gadolinium-based contrast agent; rCBV = relative cerebral blood volume; USPIO = ultrasmall superparamagnetic iron oxide.
From Farrell et al. 2013, used with permission from Neurology.
Future applications (ultrahigh field MRI, seizure imaging, functional MRI and vascular imaging in the CNS) can be found in the online supplement.
Body imaging with ferumoxytol
Liver and spleen
Metastases are the most common solid liver lesions (though benign hepatic hemangiomas are also frequent) and MRI is considered the definitive tool for differentiating these entities in the oncologic setting. However, this task can be difficult when lesions are small or when hemangiomas are atypical or of the sclerosing subtype. In many cases, small hemangiomas can be differentiated from other lesions based on retention of intravascular contrast agents such as gadofosveset trisodium or ferumoxytol on delayed scans (57, 58).
Significant negative (hypointense) T2 enhancement of normal liver parenchyma is seen within 10 min of IV ferumoxytol administration. Since ferumoxytol is taken up by the MPS, it has the potential to serve as a tool for measuring MPS dysfunction. Quantitative measures of MPS function, as estimated from dynamic imaging of ferumoxytol uptake in the liver, may prove to be useful predictors of graft dysfunction and rejection, although validations in humans are still pending (59).
Pancreas
CT is the imaging method of choice for pancreatic diseases including pancreatic adenocarcinoma, pancreatitis and neuroendocrine tumors of the pancreas. Imaging of the pancreas with MRI holds significant promise secondary to the many inherent contrast mechanisms, particularly with novel contrast agents.
Blood vessel density is known to be markedly lower in pancreatic ductal adenocarcinoma (PDAC) compared with other malignancies (60), which may explain its poor response to anti-angiogenic therapies, and makes PDAC a poor choice for interrogation with dynamic techniques. Because of the long intravascular blood pool residence time, iron oxide nanoparticles offer a steady-state solution for precise measurements of microvascular parameters as demonstrated in preclinical studies (61–64) and are better imaging agents for PDAC. In subcutaneous xenograft models, MRI of magnetic nanoparticles provides a non-invasive, accurate assessment of fractional blood volume and vessel size index (62–72).
The delayed uptake of magnetic nanoparticles by macrophages (73) has also been a focus of MRI for multiple applications in the pancreas as well, including improved delineation of pancreatic adenocarcinoma (17) and for quantifying inflammation in patients with early onset type I diabetes (Figure 11) (74, 75).
Figure 11.
Increased pancreatic nanoparticle accumulation in patients with type 1 diabetes (T1D). Single-slice (upper row) and 3D volume sets (lower row) of a representative patient with recently diagnosed T1D (left) and a normal control subject. From Gaglia et al., 2015 used with permission of PNAS.
Staging of malignant tumors
The current anatomic imaging techniques for nodal staging (e.g. CT, MRI) rely on morphologic lymph node characteristics such as size, shape and morphology, but sensitivity for differentiating malignant from benign lymph nodes remains low (76). Sensitivity could be improved using USPIO, however, most of the available data have been obtained with USPIOs other than ferumoxytol (77, 78). Iron oxide particles are taken up by and retained in normal lymph nodes, resulting in signal loss on T2- and T2*- weighted images (Figure 12) (79). When nodes are infiltrated with malignant cells, nodal USPIO nanoparticle uptake capacity reduces and malignant nodes retain high signal intensity on T2*-weighted images. Optimal node T2*-weighted imaging contrast with ferumoxytol can be achieved 24–48 h post-injection (Figures 12 and 13) (16, 79).
Figure 12.
Benign (A and B) and malignant (C and D) nodal patterns. Axial T2* GRE pre (A) shows intranodal high signal (white arrow) compared to hypointensity on 24 hours post (B) ferumoxytol scan, indicative of normal lymph nodes. Axial T2* GRE shows normal high precontrast intranodal signal (circle, C) compared to internal intranodal speckling (circle, D) suggesting a pathologic lymph node (black arrows indicate vascular structures). Images provided courtesy of Dr. Hamilton.
Figure 13.
Lymph nodes with an area of high signal intensity— encompassing the entire node or a large portion of it — are considered metastatic. A node with fatty hilum, complete signal void, and speckles of granularity without a definite focus of high signal intensity is considered non-metastatic. From Anzai et al., 2003 used with permission of Radiology.
Prostate
Nodal involvement is noted in 5% to 10% of patients with prostate carcinoma, but CT and MR detection sensitivity remains lower than 30% (80).
Clinical experience with ferumoxytol-enhanced MRI for mapping metastatic lymph nodes in patients with prostate cancer is limited. Harisinghani et al. (16) reported a significant drop in the SNR in benign nodes on T2*-weighted images, but little change in SNR in malignant nodes using ferumoxytol at a dose of 4 mg/kg. MRI was performed before, 5, 18, and 24 hours after ferumoxytol injection. The most appropriate dose of ferumoxytol is uncertain as lymph nodes in the pelvis are heterogeneous. Turkbey et al. found that a higher dose of IV ferumoxytol (up to 7.5 mg/kg) is needed to completely darken normal pelvic nodes compared to ferumoxtran-10 (81).
Direct injection of ferumoxytol into the prostate (lymphography) has shown promise in mapping sentinel lymph nodes, as demonstrated in non-human primates (82).
Other potential applications in body imaging (breast and sentinel lymph node, colorectal-, adrenal gland MRI and stem cell labeling) can be found in the online supplement.
Inflammation
Focal inflammation can mimic neoplasia. By labeling macrophages with ferumoxytol in vivo, macrophage trafficking can be detected and inflammatory lesions can be localized (83, 84).
Ferumoxytol-enhanced MRI is promising for assessing rheumatologic diseases and differentiating acute from chronic inflammatory kidney disease (85–88). Ferumoxytol-enhanced MRI has also been used for diagnosing osteomyelitis in the feet of patients with diabetes (Figure 14) (89) as well as detecting the activity of Crohn’s disease (90).
Figure 14.
Second metatarsal osteomyelitis in a 25-year-old patient with a history of a shotgun injury on the foot. T1-weighted noncontrast MR (A) shows hypointense signal involving the 2nd metatarsal (red arrow) and marked enhancement on gadolinium-enhanced T1-FS MR (B).. Noncontrast T2-weighted MR shows edema in the 2nd metatarsal (red arrow) and surrounding soft tissues (C. T2-weighted MR obtained 14 h after ferumoxytol administration (D) shows marked hypointensity compatible with ferumoxytol uptake, and consistent with the diagnosis of osteomyelitis. From A Neuwelt et al., 2016 used with permission of JMRI.
Pediatric MR imaging with ferumoxytol
Using ferumoxytol as an MR imaging agent in the pediatric population has similar potential benefits as in adults. Specific advantages include separation of IV cannula placement/ferumoxytol administration from the MRI scanning itself (which may improve the cooperation of the child if MRI is performed without anesthesia) (32). For pediatric brain tumor patients, undergoing MRI with anesthesia, gadolinium and ferumoxytol in a single imaging session was well tolerated (33). 3D contrast enhanced MRA applications in pediatric patients with CKD have been reported to evaluate IV access placement, vascular thrombosis, cardiac and renal transplantation anastomosis, re-transplantation or biliary/liver dysfunction, eliminating concerns about nephrogenic systemic fibrosis (30, 91). Finally, ferumoxytol may provide a higher level of confidence for the delineation of small vessels prior to surgery in body MRA (32).
Many types of lymphomas, soft tissue sarcomas and bone sarcomas are staged with 18F-FDG-PET/CT (92). In order to provide a radiation free alternative, whole body diffusion-weighted MR (WB-DW MR) imaging has been developed without the use of contrast agents. However, the use of ferumoxytol with this technique may be advantageous(Figure 15) (28).
Figure 15.
Comparison of ionizing-radiation-free WB-DW MRI scans and 18F-FDG PET/CT scans for the detection of malignant lymphoma. (A) Axial WB-DW MRI and (B) 18F-FDG PET/CT scan of a 15-year-old patient with stage IIIA Hodgkin’s lymphoma shows positive retroperitoneal lymph nodes (arrows). (C) Axial WB-DW MRI and (D) 18F-FDG PET/CT scan of a 14-year-old patient with class IIB Hodgkin’s lymphoma shows positive right inguinal lymph nodes (arrows). Integrated ferumoxytol-enhanced WB-DW MR images provided equal sensitivities and specificities for cancer staging compared to 18F-FDG-PET/ CT (28). More recent developments marry advantages of both imaging technologies towards 18F-FDG- PET/MR applications wherein ferumoxytol-enhanced anatomical T1-weighted MR images are merged with 18F-FDG-PET images (112). On 18F-FDG-PET/MR scans, ferumoxytol can help to differentiate malignant lymph nodes (18F-FDG positive, no iron uptake) from benign, inflammatory nodes (18F-FDG positive and iron uptake) as well as neoplastic bone marrow disease (18F-FDG positive, no iron uptake) from normal hypercellular hematopoietic marrow (18F-FDG positive and iron uptake) WB-DW=whole-body diffusion-weighted. 18F-FDG=18F-fluorodeoxyglucose. From Klenk et al., 2014 used with permission of The Lancet Oncology.
Vascular MR imaging with ferumoxytol
For contrast enhanced MRA, MR users have traditionally employed timed, first pass imaging of a gadolinium bolus, focused on the arterial or venous territory of interest. For the majority of GBCA, the volume of distribution is the extracellular fluid space and this is assessed quickly after the arterial first pass. The time window for a pure or predominantly vascular phase is therefore very short and the acquisition can be technically challenging. Gadofosveset is the one GBCA with an extended blood residence time due to reversible binding to serum albumin. However, compared to ferumoxytol, gadofosveset has a much shorter intravascular half-life (30 min vs 14–21 h) and at any given time, ~20% is distributed in the extracellular fluid space. Moreover, gadofosveset is being withdrawn from production such that by the end of 2016, ferumoxytol will be the only intravascular MRI contrast agent available for human use in the U.S.
Patients with vascular disease are a challenging group for imaging; many (20–30%) are diabetic with associated difficult vein access (93) and many (20–40%) have CKD, which can prohibit the use of iodine-and gadolinium-based contrast agents (34, 94). Ferumoxytol is a very attractive option in these patients and it can be used without the need for bolus timing or complex image acquisition schemes. Ferumoxytol’s excellent safety profile has already been established in patients with chronic renal insufficiency, though it must always be used with close physiological monitoring.
In recent years, there have been several reports of the successful use of ferumoxytol for vascular imaging in adults and children with renal failure (30, 32, 34, 57, 91, 95, 96). The studies suggest that ferumoxytol provides similar or improved quality images compared to GBCAs, without the concerns of long term gadolinium accumulation (1–4). Some of the potential applications for ferumoxytol in vascular imaging are summarized in Table 3.
Table 3.
Applications, advantages and the disadvantages of ferumoxytol enhanced MRI in patients with vascular diseases
| Region | Possibilities | Advantages | Disadvantages |
|---|---|---|---|
| All modalities and general benefits and drawbacks | MRA and MRV Vessel wall visualization First-pass and steady-state images Whole body imaging |
No ionizing radiation Nontoxic contrast agent Non-invasive Multiple planes Long imaging window (blood pool agent) Shows inflammation Highly reproducible |
Close physiological monitoring necessary |
| Carotid and intracerebral arteries | Carotid CE-MRA - vascular integrity assessment - stenosis assessment - complete plaque morphology evaluation Circle of Willis (CoW) CE-MRA CoW evaluation for treatment planning Silent infarction demonstration |
Direct visualization of the vessel wall Excellent soft-tissue contrast for plaque analysis More accurate in predicting degree of stenosis than DSA Plaque follow-up, progression monitoring |
Close physiological monitoring necessary Not suitable for visualizing calcification |
| Aorta | Aneurysm Dissection Coarctation Endoleak Infective diseases diagnosis |
Visualization of wall inflammation More sensitive for post EVAR endoleaks than CTA |
Close physiological monitoring necessary |
| Cardiac | Coronary artery visualization Detection and remodeling of myocardial infarct Inflammatory diseases (myocarditis) Congenital heart diseases Chronic heart failure measurements Acute transplant rejection |
High resolution and 3D images of the cardiac chambers and thoracic vessels Global and regional contractile function assessment Blood flow measurements High sensitivity and specificity for acute inflammation Response monitoring for treatment No breath holding Shorter acquisition time |
Close physiological monitoring necessary Stents and metallic artifacts |
| Lower extremities | Lower limb CE-MRA Possibility for blood volume mapping Revascularization follow up |
Post-revascularization increased blood volume can be shown | Difficulty in distinguishing arteries and veins on steady-state images |
| Visceral arteries | Renal artery evaluation Kidney transplant follow up Mesenteric ischemia Splenic artery aneurysm |
Beneficial in any stage of renal insufficiency | |
| Veins | Vena cava and iliac vein thrombosis diagnosis and follow up Agenesis and chronic occlusion Vein mapping prior to dialysis graft placement |
The whole abdominopelvic deep venous system is assessable Pulmonary thromboembolism can be visualized (in major vessels) Better sensitivity for thrombus imaging than non-contrast techniques No bolus needed Long imaging window Higher resolution Fewer motion artifacts High SNR |
|
| Pediatric | Congenital heart disease Vascular malformations Kidney diseases and transplantation follow up |
Whole body imaging is available | Sedation or anesthesia may be needed |
MRA: magnetic resonance angiography. MRV: magnetic resonance venography. CE-MRA: contrast-enhanced MRA. CoW: Circle of Willis. DSA: digital subtraction angiography. EVAR: endovascular aneurysm repair. CTA: computed tomography angiography. SNR: signal to noise ratio.
Cardiac imaging
To date, there have been very few reports of the use of ferumoxytol in patients with ischemic heart disease. In patients with acute myocardial infarction, Alam et al. (97) and Yilmaz et al. (98) showed macrophage activation in injured myocardium. These authors exploited the effects of ferumoxytol in causing signal loss due to its effect on T2 values and did not address vascular enhancement due to shortening of T1 values.
In congenital heart disease, accurate anatomical assessment is important for surgical or interventional planning. Conventional first pass MRA and 2D cardiac cine methods provide limited definition of intracardiac structures. On the other hand, ferumoxytol supports the acquisition of 4-dimensional images of the beating heart with Four-Dimensional, Multiphase, Steady-State Imaging with Contrast Enhancement (MUSIC) (Figure 16) (99). Similarly, comprehensive motion-compensated highly accelerated 4D flow MRI with ferumoxytol enhances the quality of 4-dimensional flow information with reduced respiratory motion artifacts in children with congenital heart disease (100, 101).
Figure 16.

Kawasaki Disease. Multi-lobed Aneurysms (arrows) of the Right and Left Coronary Arteries in a 3 year-old patient. Single frame from a MUSIC acquisition (99). Systemic arteries are colored red. Images provided courtesy of Dr. Finn.
The imaging goals for most congenital heart diseases include quantification of blood flow in the great vessels, determination of cardiac chamber volumes and contractility, and assessment of segmental anatomy. Typically, this requires a lengthy MRI scan with over an hour of repeated breath holding under anesthesia with specialized technologists and physicians on hand to prescribe customized planes for image acquisition. A compelling advantage of ferumoxytol is that it enables volumetric temporally-resolved high resolution imaging without breath holding (99). Further, a free-breathing volumetric comprehensive MRI yielding flow, function, and anatomic assessment in less than 10 min has been achieved (100). Additionally, ventricular mass may be quantified with this same technique (101). As a result, no special operator knowledge of cardiac anatomy is required, and the duration of anesthesia and complexity of the procedure for congenital heart MRI can be greatly reduced. These techniques stand to revolutionize the approach to MR imaging of children with complex congenital heart diseases.
Aortic imaging
Aortic imaging can be performed with a computed tomography angiography (CTA), non-contrast MRI or gadolinium enhanced MRA. Blood volume, flow measurements, and morphological assessments can be made with contrast enhanced MRA using a combination of time-resolved and steady-state images. Inflammation in the aortic wall can potentially be assessed using in vivo macrophage labeling with ferumoxytol, which is not available with gadolinium-enhanced imaging. Inflammation plays a key role in the progression and vulnerability to rupture of atherosclerotic plaques in many arterial territories. The presence of inflammation can also predict vessel patency (102, 103).
Additionally, due to its long intravascular half-life, ferumoxytol can be used for the detection of aortic endoleaks after endovascular aneurysm repair. A systematic review showed that MRA detected almost twice as many endoleaks as CTA (104). Iron oxide-enhanced MRA (either with ferumoxytol or with ferucarbotran) may be advantageous for demonstrating endoleaks (105, 106).
Visceral arteries and arteriovenous (AV) fistulas
Patients with end-organ ischemia in the abdominal vasculature often have diabetes and renal insufficiency. Ferumoxytol may therefore be a useful agent for assessing renal artery stenosis, pre- and post-transplantation vessel patency, and for new venous access. In pre and post renal and liver transplantation patients, ferumoxytol has been used to assess vascular integrity (30, 95). The frequent occurrence of venous occlusion in these patients, and the requirement to confirm or re-establish venous access, makes high resolution venous imaging with ferumoxytol invaluable (Figure 17) (91).
Figure 17.
Volume rendered reconstruction of an inferior vena cava occlusion in a 58-year-old male with chronic renal failure. Steady-state, breath held, high resolution MRA with ferumoxytol at 3.0 T. Huge pelvic and abdominal wall collaterals are clearly visualized, as well as a dilated azygous vein. Blue: systemic veins; Purple: portal vessels purple; Red: systemic arteries. Images provided courtesy of Dr. Finn.
Most studies regarding pre-fistula assessments involve pediatric patients, since this group benefits the most from ferumoxytol-enhanced MRA. In a feasibility study using ferumoxytol for clinical pediatric cardiovascular imaging, Ruanwattanapaisarn et al. evaluated renal transplant or post-transplant complications, vascular shunts, stenosis, aneurysms and congenital heart disease, and found excellent image quality of the hepatic arteries, superior mesenteric artery, renal arteries, pulmonary arteries, pulmonary veins, valves and ventricles (32). Ferumoxytol-enhanced MRA provided consistently superior quality TOF MRA images in patients with AV fistulas (96).
Other applications in vascular imaging (carotid-, peripheral artery imaging and magnetic resonance venography) can be found in the online supplement.
Discussion
In this review, we draw attention to ferumoxytol as a contrast agent for cross sectional imaging in patients with renal failure and highlighted the variety of clinical indications in which ferumoxytol may not only be an alternative to GBCA, but also a specialized contrast agent with unique properties. These include the extended blood pool phase, and uptake into the MPS in the delayed phase, where nanoparticles are metabolized as iron.
Ferumoxytol has been approved by the FDA for iron replacement in 2009, and it is available for off label clinical use. A large and growing body of literature has been reviewed in this manuscript showing potentials of ferumoxytol imaging. Although most available evidence is based on single center studies, pilot studies, retrospective analyses or preclinical results, there are new multicenter, multiplatform studies being designed (clinicaltrials.gov, NCT02359097). Application for FDA approval of using ferumoxytol as an imaging agent is in preparation, which would further ease its widespread use of ferumoxytol in MR imaging.
Limitations
Besides known hypersensitivity or iron metabolic disorders, there is no absolute contraindication of ferumoxytol for MR imaging, and importantly is safe in patients with renal impairment. Vascular visualization improves early after administration, while late enhancement visualizes parenchymal/intracellular enhancement, which may require an additional visit, posing a logistical limitation with MRI scheduling. Signal change in the brain may persist from a few days to a week. Uptake in the liver, spleen and bone marrow may alter MRI signal for months, therefore, radiologists must be aware of prior history of IV iron oxide use. While hypersensitivity reactions occur, the incidence of serious ferumoxytol rela1ted hypersensitivity is very low and new FDA guidelines aim to further improve patient safety.
Conclusion
In conclusion, there are numerous potential applications of ferumoxytol for MRI throughout the body, including the CNS, various organs outside of the CNS, and the cardiovascular system. Whether used where GBCA is contraindicated, or for applications that are currently outside the scope of GBCA, ferumoxytol has shown sufficient promise to warrant vigorous research into its ultimate clinical role. Radiological modalities which are noninvasive, radiation free and safe are generally preferred and should be further developed; ferumoxytol MRI may be an important step in this direction.
Supplementary Material
Figure 10.
Ferumoxytol MR angiography improves visualization of cerebral microvasculature when compared to Gadolinium at 7T field strength. Gadolinium-enhanced 3D T1-weighted gradient echo volumetric interpolated brain examination (VIBE) MRA of the supraclinoid internal carotid arteries (left) shows faint visualization of lenticulostriate vessels (white arrows). Ferumoxytol-enhanced MRA (right) demonstrates markedly improved visualization of the lenticulostriate microvasculature (white arrows). Images provided courtesy of Dr. Barajas.
Acknowledgments
Funding:
This work was supported in part by National Institutes of Health grants NS053468 and CA137488-15S1, in part with Federal funds from the National Cancer Institute, under Contract No. HHSN261200800001E, a Veterans Administration Merit Review Grant, and by the Walter S. and Lucienne Driskill Foundation, all to EAN; R01 HL127153 IND 129441 (Feraheme as an MRI Contrast Agent for Pediatric Congenital Heart Disease - on clinical trials.gov) to JPF; and 2R01AR054458, R01 HD081123A, R21CA176519, R21AR066302, R21CA190196 to HED.
Footnotes
Authors Conflicts of Interest:
The authors have no conflicts of interest to disclose.
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References
- 1.Kanda T, Oba H, Toyoda K, Kitajima K, Furui S. Brain gadolinium deposition after administration of gadolinium-based contrast agents. Japanese journal of radiology. 2016;34(1):3–9. doi: 10.1007/s11604-015-0503-5. [DOI] [PubMed] [Google Scholar]
- 2.Ramalho J, Semelka RC, AlObaidy M, Ramalho M, Nunes RH, Castillo M. Signal intensity change on unenhanced T1-weighted images in dentate nucleus following gadobenate dimeglumine in patients with and without previous multiple administrations of gadodiamide. European radiology. 2016 doi: 10.1007/s00330-016-4269-7. [DOI] [PubMed] [Google Scholar]
- 3.Reeder SB, Gulani V. Gadolinium Deposition in the Brain: Do We Know Enough to Change Practice? Radiology. 2016;279(1):323–6. doi: 10.1148/radiol.2016151372. [DOI] [PubMed] [Google Scholar]
- 4.Stojanov D, Aracki-Trenkic A, Benedeto-Stojanov D. Gadolinium deposition within the dentate nucleus and globus pallidus after repeated administrations of gadolinium-based contrast agents-current status. Neuroradiology. 2016;58(5):433–41. doi: 10.1007/s00234-016-1658-1. [DOI] [PubMed] [Google Scholar]
- 5.Kowalczyk M, Banach M, Rysz J. Ferumoxytol: a new era of iron deficiency anemia treatment for patients with chronic kidney disease. Journal of nephrology. 2011;24(6):717–22. doi: 10.5301/jn.5000025. [DOI] [PubMed] [Google Scholar]
- 6.Balakrishnan VS, Rao M, Kausz AT, Brenner L, Pereira BJ, Frigo TB, et al. Physicochemical properties of ferumoxytol, a new intravenous iron preparation. European journal of clinical investigation. 2009;39(6):489–96. doi: 10.1111/j.1365-2362.2009.02130.x. [DOI] [PubMed] [Google Scholar]
- 7.Hetzel D, Strauss W, Bernard K, Li Z, Urboniene A, Allen LF. A Phase III, randomized, open-label trial of ferumoxytol compared with iron sucrose for the treatment of iron deficiency anemia in patients with a history of unsatisfactory oral iron therapy. American journal of hematology. 2014;89(6):646–50. doi: 10.1002/ajh.23712. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.Macdougall IC, Strauss WE, McLaughlin J, Li Z, Dellanna F, Hertel J. A randomized comparison of ferumoxytol and iron sucrose for treating iron deficiency anemia in patients with CKD. Clinical journal of the American Society of Nephrology : CJASN. 2014;9(4):705–12. doi: 10.2215/CJN.05320513. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Vadhan-Raj S, Ford DC, Dahl NV, Bernard K, Li Z, Allen LF, et al. Safety and efficacy of ferumoxytol for the episodic treatment of iron deficiency anemia in patients with a history of unsatisfactory oral iron therapy: Results of a phase III, open-label, 6-month extension study. American journal of hematology. 2016;91(2):E3–5. doi: 10.1002/ajh.24240. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Gahramanov S, Muldoon LL, Varallyay CG, Li X, Kraemer DF, Fu R, et al. Pseudoprogression of glioblastoma after chemo- and radiation therapy: diagnosis by using dynamic susceptibility-weighted contrast-enhanced perfusion MR imaging with ferumoxytol versus gadoteridol and correlation with survival. Radiology. 2013;266(3):842–52. doi: 10.1148/radiol.12111472. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.FDA Drug Safety Communication. FDA strengthens warnings and changes prescribing instructions to decrease the risk of serious allergic reactions with anemia drug Feraheme (ferumoxytol) 2015 [Google Scholar]
- 12.Christen T, Ni W, Qiu D, Schmiedeskamp H, Bammer R, Moseley M, et al. High-resolution cerebral blood volume imaging in humans using the blood pool contrast agent ferumoxytol. Magnetic resonance in medicine. 2013;70(3):705–10. doi: 10.1002/mrm.24500. [DOI] [PubMed] [Google Scholar]
- 13.Varallyay CG, Nesbit E, Fu R, Gahramanov S, Moloney B, Earl E, et al. High-resolution steady-state cerebral blood volume maps in patients with central nervous system neoplasms using ferumoxytol, a superparamagnetic iron oxide nanoparticle. J Cereb Blood Flow Metab. 2013;33(5):780–6. doi: 10.1038/jcbfm.2013.36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Finn JP, Nguyen KL, Han F, Zhou Z, Salusky I, Ayad I, et al. Cardiovascular MRI with ferumoxytol. Clinical radiology. 2016;71(8):796–806. doi: 10.1016/j.crad.2016.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Neuwelt EA, Varallyay CG, Manninger S, Solymosi D, Haluska M, Hunt MA, et al. The potential of ferumoxytol nanoparticle magnetic resonance imaging, perfusion, and angiography in central nervous system malignancy: a pilot study. Neurosurgery. 2007;60(4):601–11. doi: 10.1227/01.NEU.0000255350.71700.37. discussion 11–2. [DOI] [PubMed] [Google Scholar]
- 16.Harisinghani M, Ross RW, Guimaraes AR, Weissleder R. Utility of a new bolus-injectable nanoparticle for clinical cancer staging. Neoplasia (New York, NY) 2007;9(12):1160–5. doi: 10.1593/neo.07940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Hedgire S, Mino-Kenudson M, Elmi A, Thayer S, Fernandez-del Castillo C, Harisinghani M. Enhanced primary tumor delineation in pancreatic adenocarcinoma using ultrasmall super paramagnetic iron oxide nanoparticle-ferumoxytol: an initial experience with histopathologic correlation. Int J Nanomedicine. 2014;9:1891–6. doi: 10.2147/IJN.S59788. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Daldrup-Link HE, Golovko D, Ruffell B, Denardo DG, Castaneda R, Ansari C, et al. MRI of tumor-associated macrophages with clinically applicable iron oxide nanoparticles. Clin Cancer Res. 2011;17(17):5695–704. doi: 10.1158/1078-0432.CCR-10-3420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.McConnell HL, Schwartz DL, Richardson BE, Woltjer RL, Muldoon LL, Neuwelt EA. Ferumoxytol nanoparticle uptake in brain during acute neuroinflammation is cell-specific. Nanomedicine : nanotechnology, biology, and medicine. 2016 doi: 10.1016/j.nano.2016.03.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Muehe AM, Feng D, von Eyben R, Luna-Fineman S, Link MP, Muthig T, et al. Safety Report of Ferumoxytol for Magnetic Resonance Imaging in Children and Young Adults. Investigative radiology. 2016;51(4):221–7. doi: 10.1097/RLI.0000000000000230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Provenzano R, Schiller B, Rao M, Coyne D, Brenner L, Pereira BJ. Ferumoxytol as an intravenous iron replacement therapy in hemodialysis patients. Clin J Am Soc Nephrol. 2009;4(2):386–93. doi: 10.2215/CJN.02840608. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Singh A, Patel T, Hertel J, Bernardo M, Kausz A, Brenner L. Safety of ferumoxytol in patients with anemia and CKD. American journal of kidney diseases : the official journal of the National Kidney Foundation. 2008;52(5):907–15. doi: 10.1053/j.ajkd.2008.08.001. [DOI] [PubMed] [Google Scholar]
- 23.Spinowitz BS, Kausz AT, Baptista J, Noble SD, Sothinathan R, Bernardo MV, et al. Ferumoxytol for treating iron deficiency anemia in CKD. Journal of the American Society of Nephrology : JASN. 2008;19(8):1599–605. doi: 10.1681/ASN.2007101156. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Schiller B, Bhat P, Sharma A. Safety and effectiveness of ferumoxytol in hemodialysis patients at 3 dialysis chains in the United States over a 12-month period. Clinical therapeutics. 2014;36(1):70–83. doi: 10.1016/j.clinthera.2013.09.028. [DOI] [PubMed] [Google Scholar]
- 25.Bashir MR, Mody R, Neville A, Javan R, Seaman D, Kim CY, et al. Retrospective assessment of the utility of an iron-based agent for contrast-enhanced magnetic resonance venography in patients with endstage renal diseases. Journal of magnetic resonance imaging : JMRI. 2014;40(1):113–8. doi: 10.1002/jmri.24330. [DOI] [PubMed] [Google Scholar]
- 26.D’Arceuil H, Coimbra A, Triano P, Dougherty M, Mello J, Moseley M, et al. Ferumoxytol enhanced resting state fMRI and relative cerebral blood volume mapping in normal human brain. NeuroImage. 2013;83:200–9. doi: 10.1016/j.neuroimage.2013.06.066. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Hasan DM, Amans M, Tihan T, Hess C, Guo Y, Cha S, et al. Ferumoxytol-enhanced MRI to Image Inflammation within Human Brain Arteriovenous Malformations: A Pilot Investigation. Translational stroke research. 2012;3(Suppl 1):166–73. doi: 10.1007/s12975-012-0172-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Klenk C, Gawande R, Uslu L, Khurana A, Qiu D, Quon A, et al. Ionising radiation-free whole-body MRI versus (18)F-fluorodeoxyglucose PET/CT scans for children and young adults with cancer: a prospective, non-randomised, single-centre study. Lancet Oncol. 2014;15(3):275– 85. doi: 10.1016/S1470-2045(14)70021-X. [DOI] [PubMed] [Google Scholar]
- 29.Li W, Tutton S, Vu AT, Pierchala L, Li BS, Lewis JM, et al. First-pass contrast-enhanced magnetic resonance angiography in humans using ferumoxytol, a novel ultrasmall superparamagnetic iron oxide (USPIO)-based blood pool agent. Journal of magnetic resonance imaging : JMRI. 2005;21(1):46–52. doi: 10.1002/jmri.20235. [DOI] [PubMed] [Google Scholar]
- 30.Nayak AB, Luhar A, Hanudel M, Gales B, Hall TR, Finn JP, et al. High-resolution, whole-body vascular imaging with ferumoxytol as an alternative to gadolinium agents in a pediatric chronic kidney disease cohort. Pediatric nephrology (Berlin, Germany) 2015;30(3):515–21. doi: 10.1007/s00467-014-2953-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ning P, Zucker EJ, Wong P, Vasanawala SS. Hemodynamic safety and efficacy of ferumoxytol as an intravenous contrast agents in pediatric patients and young adults. Magnetic resonance imaging. 2016;34(2):152–8. doi: 10.1016/j.mri.2015.10.019. [DOI] [PubMed] [Google Scholar]
- 32.Ruangwattanapaisarn N, Hsiao A, Vasanawala SS. Ferumoxytol as an off-label contrast agent in body 3T MR angiography: a pilot study in children. Pediatric radiology. 2015;45(6):831– 9. doi: 10.1007/s00247-014-3226-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Thompson EM, Guillaume DJ, Dosa E, Li X, Nazemi KJ, Gahramanov S, et al. Dual contrast perfusion MRI in a single imaging session for assessment of pediatric brain tumors. Journal of neuro-oncology. 2012;109(1):105–14. doi: 10.1007/s11060-012-0872-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Walker JP, Nosova E, Sigovan M, Rapp J, Grenon MS, Owens CD, et al. Ferumoxytol-enhanced magnetic resonance angiography is a feasible method for the clinical evaluation of lower extremity arterial disease. Ann Vasc Surg. 2015;29(1):63–8. doi: 10.1016/j.avsg.2014.09.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Vasanawala SS, Nguyen KL, Hope MD, Bridges MD, Hope TA, Reeder SB, et al. Safety and technique of ferumoxytol administration for MRI. Magnetic resonance in medicine. 2016;75(5):2107–11. doi: 10.1002/mrm.26151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Hamilton BE, Nesbit GM, Dosa E, Gahramanov S, Rooney B, Nesbit EG, et al. Comparative analysis of ferumoxytol and gadoteridol enhancement using T1- and T2-weighted MRI in neuroimaging. AJR American journal of roentgenology. 2011;197(4):981–8. doi: 10.2214/AJR.10.5992. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Dosa E, Guillaume DJ, Haluska M, Lacy CA, Hamilton BE, Njus JM, et al. Magnetic resonance imaging of intracranial tumors: intra-patient comparison of gadoteridol and ferumoxytol. Neuro Oncol. 2011;13(2):251–60. doi: 10.1093/neuonc/noq172. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Hamilton BEN, GM, Barajas R, Ambrady P, Neuwelt EA, editors. Ferumoxytol enhanced MRI detection of brain metastatic disease. ASNR; Washington DC: 2016. [Google Scholar]
- 39.Aghighi M, Golovko D, Ansari C, Marina NM, Pisani L, Kurlander L, et al. Imaging Tumor Necrosis with Ferumoxytol. PloS one. 2015;10(11):e0142665. doi: 10.1371/journal.pone.0142665. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Dosa E, Tuladhar S, Muldoon LL, Hamilton BE, Rooney WD, Neuwelt EA. MRI using ferumoxytol improves the visualization of central nervous system vascular malformations. Stroke. 2011;42(6):1581–8. doi: 10.1161/STROKEAHA.110.607994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Neuwelt EA, Hamilton BE, Varallyay CG, Rooney WR, Edelman RD, Jacobs PM, et al. Ultrasmall superparamagnetic iron oxides (USPIOs): a future alternative magnetic resonance (MR) contrast agent for patients at risk for nephrogenic systemic fibrosis (NSF)? Kidney Int. 2009;75(5):465–74. doi: 10.1038/ki.2008.496. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Hamilton BE, Woltjer RL, Prola-Netto J, Nesbit GM, Gahramanov S, Pham T, et al. Ferumoxytol-enhanced MRI differentiation of meningioma from dural metastases: a pilot study with immunohistochemical observations. J Neurooncol. 2016 doi: 10.1007/s11060-016-2175-0. [DOI] [PubMed] [Google Scholar]
- 43.Abdoli M, Freedman MS. Neuro-oncology dilemma: Tumour or tumefactive demyelinating lesion. Multiple sclerosis and related disorders. 2015;4(6):555–66. doi: 10.1016/j.msard.2015.07.013. [DOI] [PubMed] [Google Scholar]
- 44.Farrell BT, Hamilton BE, Dosa E, Rimely E, Nasseri M, Gahramanov S, et al. Using iron oxide nanoparticles to diagnose CNS inflammatory diseases and PCNSL. Neurology. 2013;81(3):256–63. doi: 10.1212/WNL.0b013e31829bfd8f. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Law M, Yang S, Wang H, Babb JS, Johnson G, Cha S, et al. Glioma grading: sensitivity, specificity, and predictive values of perfusion MR imaging and proton MR spectroscopic imaging compared with conventional MR imaging. AJNR American journal of neuroradiology. 2003;24(10):1989–98. [PMC free article] [PubMed] [Google Scholar]
- 46.Danchaivijitr N, Waldman AD, Tozer DJ, Benton CE, Brasil Caseiras G, Tofts PS, et al. Low-grade gliomas: do changes in rCBV measurements at longitudinal perfusion-weighted MR imaging predict malignant transformation? Radiology. 2008;247(1):170–8. doi: 10.1148/radiol.2471062089. [DOI] [PubMed] [Google Scholar]
- 47.Brandsma D, Stalpers L, Taal W, Sminia P, van den Bent MJ. Clinical features, mechanisms, and management of pseudoprogression in malignant gliomas. Lancet Oncol. 2008;9(5):453–61. doi: 10.1016/S1470-2045(08)70125-6. [DOI] [PubMed] [Google Scholar]
- 48.Nasseri M, Gahramanov S, Netto JP, Fu R, Muldoon LL, Varallyay C, et al. Evaluation of pseudoprogression in patients with glioblastoma multiforme using dynamic magnetic resonance imaging with ferumoxytol calls RANO criteria into question. Neuro Oncol. 2014;16(8):1146–54. doi: 10.1093/neuonc/not328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Varallyay CG, Muldoon LL, Gahramanov S, Wu YJ, Goodman JA, Li X, et al. Dynamic MRI using iron oxide nanoparticles to assess early vascular effects of antiangiogenic versus corticosteroid treatment in a glioma model. J Cereb Blood Flow Metab. 2009;29(4):853–60. doi: 10.1038/jcbfm.2008.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Cohen JV, Alomari AK, Vortmeyer AO, Jilaveanu LB, Goldberg SB, Mahajan A, et al. Melanoma Brain Metastasis Pseudoprogression after Pembrolizumab Treatment. Cancer Immunol Res. 2016;4(3):179–82. doi: 10.1158/2326-6066.CIR-15-0160. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Hodi FS, Hwu WJ, Kefford R, Weber JS, Daud A, Hamid O, et al. Evaluation of Immune- Related Response Criteria and RECIST v1.1 in Patients With Advanced Melanoma Treated With Pembrolizumab. J Clin Oncol. 2016;34(13):1510–7. doi: 10.1200/JCO.2015.64.0391. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Okada H, Weller M, Huang R, Finocchiaro G, Gilbert MR, Wick W, et al. Immunotherapy response assessment in neuro-oncology: a report of the RANO working group. Lancet Oncol. 2015;16(15):e534–42. doi: 10.1016/S1470-2045(15)00088-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Gahramanov S, Varallyay C, Tyson RM, Lacy C, Fu R, Netto JP, et al. Diagnosis of pseudoprogression using MRI perfusion in patients with glioblastoma multiforme may predict improved survival. CNS Oncol. 2014;3(6):389–400. doi: 10.2217/cns.14.42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Vellinga MM, Oude Engberink RD, Seewann A, Pouwels PJ, Wattjes MP, van der Pol SM, et al. Pluriformity of inflammation in multiple sclerosis shown by ultra-small iron oxide particle enhancement. Brain : a journal of neurology. 2008;131(Pt 3):800–7. doi: 10.1093/brain/awn009. [DOI] [PubMed] [Google Scholar]
- 55.Tagge A Comparative Volumetric Study in Acute Japanese Macaque Encephalomyelitis. Proc Int Soc Magn Reson Med. 2015 [Google Scholar]
- 56.Axthelm MK, Bourdette DN, Marracci GH, Su W, Mullaney ET, Manoharan M, et al. Japanese macaque encephalomyelitis: a spontaneous multiple sclerosis-like disease in a nonhuman primate. Annals of neurology. 2011;70(3):362–73. doi: 10.1002/ana.22449. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Bashir MR, Bhatti L, Marin D, Nelson RC. Emerging applications for ferumoxytol as a contrast agent in MRI. Journal of magnetic resonance imaging : JMRI. 2015;41(4):884–98. doi: 10.1002/jmri.24691. [DOI] [PubMed] [Google Scholar]
- 58.Milot L, Haider M, Foster L, McGregor C, Law C. Gadofosveset trisodium in the investigation of focal liver lesions in noncirrhotic liver: Early experience. Journal of magnetic resonance imaging : JMRI. 2012;36(3):738–42. doi: 10.1002/jmri.23650. [DOI] [PubMed] [Google Scholar]
- 59.Liu T, Choi H, Zhou R, Chen IW. Quantitative evaluation of the reticuloendothelial system function with dynamic MRI. PloS one. 2014;9(8):e103576. doi: 10.1371/journal.pone.0103576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Olive K, Jacobetz M, Davidson C, Gopinathan A, McIntyre D, Honess D, et al. Inhibition of Hedgehog signaling enhances delivery of chemotherapy in a mouse model of pancreatic cancer. Science. 2009;324(5933):1457–61. doi: 10.1126/science.1171362. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Boxerman J, Hamberg L, Rosen B, Weisskoff R. MR contrast due to intravascular magnetic susceptibility perturbations. Magnetic resonance in medicine. 1995;34(4):555–66. doi: 10.1002/mrm.1910340412. [DOI] [PubMed] [Google Scholar]
- 62.Bremer C, Mustafa M, Bogdanov A, Jr, Petrovsky A, Weissleder R. Steady-state blood volume measurements in Experimental tumors with different angiogenic burdens - a study in mice. Radiology. 2003;226(1):214–20. doi: 10.1148/radiol.2261012140. [DOI] [PubMed] [Google Scholar]
- 63.Dennie J, Mandeville J, Boxerman J, Packard S, Rosen B, Weisskoff R. NMR imaging of changes in vascular morphology due to tumor angiogenesis. Magnetic resonance in medicine. 1998;40:793–99. doi: 10.1002/mrm.1910400602. [DOI] [PubMed] [Google Scholar]
- 64.Guimaraes A, Rakhlin E, Weissleder R, Thayer S. Magnetic resonance imaging monitors physiological changes with antihedgehog therapy in pancreatic adenocarcinoma xenograft model. Pancreas. 2008;37(4):440–4. doi: 10.1097/MPA.0b013e31817c5113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.CTF, WSK, CDL, YRK, SJK, GD, et al. In Vivo validation of MRI vessel caliber index measurement methods with intravital optical microscopy in a u87 mouse brain tumor. Neuro Oncology. 2010 doi: 10.1093/neuonc/nop032. In Press. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Emblem K, Farrar C, Gerstner E, Batchelor T, Borra R, Rosen B, et al. Vessel caliber--a potential MRI biomarker of tumour response in clinical trials. Nat Rev Clin Oncol. 2014;11(10):566–84. doi: 10.1038/nrclinonc.2014.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Flexman J, Yung A, Yapp D, Ng S, Kozlowski P. Assessment of Vessel Size by MRI in Orthotopic Model of Human Pancreatic Cancer. Annual International IEEE EMBS Conference; 2009; Vancouver, BC, Canada. pp. 851–4. [DOI] [PubMed] [Google Scholar]
- 68.Guimaraes A, Ross R, Figuereido J, Waterman P, Weissleder R. MRI with magnetic nanoparticles monitors downstream anti-angiogenic effects of mTOR inhibition. Mol Imaging Biol. 2011;13(2):314–20. doi: 10.1007/s11307-010-0357-2. [DOI] [PubMed] [Google Scholar]
- 69.Jensen J, Chandra R. MR imaging of microvasculature. Magnetic resonance in medicine. 2000;44(2):224–30. doi: 10.1002/1522-2594(200008)44:2<224::aid-mrm9>3.0.co;2-m. [DOI] [PubMed] [Google Scholar]
- 70.Kiselev V, Strecker R, Ziyeh S, Speck O, Hennig J. Vessel size imaging in humans. Magnetic resonance in medicine. 2005;53(3):553–63. doi: 10.1002/mrm.20383. [DOI] [PubMed] [Google Scholar]
- 71.Lemasson B, Valable S, Farion R, Krainik A, Remy C, Barbier E. In vivo imaging of vessel diameter, size, and density: a comparative study between MRI and histology. Magnetic resonance in medicine. 2013;69(1):18–26. doi: 10.1002/mrm.24218. [DOI] [PubMed] [Google Scholar]
- 72.Tropres I, Grimault S, Vaeth A, Grillon E, Julien C, Payen J, et al. Vessel size imaging. Magnetic resonance in medicine. 2001;45:397–408. doi: 10.1002/1522-2594(200103)45:3<397::aid-mrm1052>3.0.co;2-3. [DOI] [PubMed] [Google Scholar]
- 73.Weissleder R, Elizaondo G, Wittenberg J, et al. Ultrasmall superparamagnetic iron oxide: characterization of a new class of contrast agents for MR imaging. Radiology. 1990;175:489–93. doi: 10.1148/radiology.175.2.2326474. [DOI] [PubMed] [Google Scholar]
- 74.Gaglia J, Guimaraes A, Harisinghani M, Turvey S, Jackson R, Benoist C, et al. Noninvasive imaging of pancreatic islet inflammation in type 1A diabetes patients. J Clin Invest. 2011;121(1):442–5. doi: 10.1172/JCI44339. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Gaglia J, Harisinghani M, Aganj I, Wojtkiewicz G, Hedgire S, Benoist C, et al. Noninvasive mapping of pancreatic inflammation in recent-onset type-1 diabetes patients. Proc Natl Acad Sci U S A. 2015;112(7):2139–44. doi: 10.1073/pnas.1424993112. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Ganeshalingam S, Koh D-M. Nodal staging. Cancer Imaging. 2009;9(1):104–11. doi: 10.1102/1470-7330.2009.0017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Barentsz J. MR imaging of pelvic lymph nodes. Cancer Imaging. 2003;3(2):130–4. [Google Scholar]
- 78.Harisinghani MG, Saini S, Slater GJ, Schnall MD, Rifkin MD. MR imaging of pelvic lymph nodes in primary pelvic carcinoma with ultrasmall superparamagnetic iron oxide (Combidex): preliminary observations. Journal of magnetic resonance imaging : JMRI. 1997;7(1):161–3. doi: 10.1002/jmri.1880070124. [DOI] [PubMed] [Google Scholar]
- 79.Anzai Y, Piccoli CW, Outwater EK, Stanford W, Bluemke DA, Nurenberg P, et al. Evaluation of neck and body metastases to nodes with ferumoxtran 10-enhanced MR imaging: phase III safety and efficacy study. Radiology. 2003;228(3):777–88. doi: 10.1148/radiol.2283020872. [DOI] [PubMed] [Google Scholar]
- 80.Wolf JS, Jr, Cher M, Dall’era M, Presti JC, Jr, Hricak H, Carroll PR. The use and accuracy of cross-sectional imaging and fine needle aspiration cytology for detection of pelvic lymph node metastases before radical prostatectomy. The Journal of urology. 1995;153(3 Pt 2):993–9. [PubMed] [Google Scholar]
- 81.Turkbey B, Agarwal HK, Shih J, Bernardo M, McKinney YL, Daar D, et al. A Phase I Dosing Study of Ferumoxytol for MR Lymphography at 3 T in Patients With Prostate Cancer. AJR American journal of roentgenology. 2015;205(1):64–9. doi: 10.2214/AJR.14.13009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Sankineni S, Smedley J, Bernardo M, Brown AM, Johnson L, Muller B, et al. Ferumoxytol as an intraprostatic MR contrast agent for lymph node mapping of the prostate: a feasibility study in non-human primates. Acta radiologica (Stockholm, Sweden : 1987) 2015 doi: 10.1177/0284185115586023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Hasan D, Chalouhi N, Jabbour P, Dumont AS, Kung DK, Magnotta VA, et al. Early change in ferumoxytol-enhanced magnetic resonance imaging signal suggests unstable human cerebral aneurysm: a pilot study. Stroke. 2012;43(12):3258–65. doi: 10.1161/STROKEAHA.112.673400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wagner S, Schnorr J, Ludwig A, Stangl V, Ebert M, Hamm B, et al. Contrast-enhanced MR imaging of atherosclerosis using citrate-coated superparamagnetic iron oxide nanoparticles: calcifying microvesicles as imaging target for plaque characterization. Int J Nanomedicine. 2013;8:767–79. doi: 10.2147/IJN.S38702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Budjan J, Neudecker S, Schock-Kusch D, Kraenzlin B, Schoenberg SO, Michaely HJ, et al. Can Ferumoxytol be Used as a Contrast Agent to Differentiate Between Acute and Chronic Inflammatory Kidney Disease?: Feasibility Study in a Rat Model. Investigative radiology. 2016;51(2):100–5. doi: 10.1097/RLI.0000000000000209. [DOI] [PubMed] [Google Scholar]
- 86.Rogers JL, Tarrant T, Kim JS. Nanoparticle-based diagnostic imaging of inflammation in rheumatic disease. Current rheumatology reviews. 2014;10(1):3–10. doi: 10.2174/1573403x10666140914154816. [DOI] [PubMed] [Google Scholar]
- 87.Simon GH, von Vopelius-Feldt J, Fu Y, Schlegel J, Pinotek G, Wendland MF, et al. Ultrasmall supraparamagnetic iron oxide-enhanced magnetic resonance imaging of antigen-induced arthritis: a comparative study between SHU 555 C, ferumoxtran-10, and ferumoxytol. Invest Radiol. 2006;41(1):45–51. doi: 10.1097/01.rli.0000191367.61306.83. [DOI] [PubMed] [Google Scholar]
- 88.Neuwelt A. Ferumoxtyol Negatively Enhances T2-weighted MRI od Pedal Osteomyelitis in vivo. Journal of magnetic resonance imaging : JMRI. 2016 doi: 10.1002/jmri.25437. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Neuwelt A, Sidhu N, Hu CA, Mlady G, Eberhardt SC, Sillerud LO. Iron-based superparamagnetic nanoparticle contrast agents for MRI of infection and inflammation. AJR American journal of roentgenology. 2015;204(3):W302–13. doi: 10.2214/AJR.14.12733. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Moy MP, Sauk J, Gee MS. The Role of MR Enterography in Assessing Crohn’s Disease Activity and Treatment Response. Gastroenterology research and practice. 2016;2016:8168695. doi: 10.1155/2016/8168695. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Luhar A, Khan S, Finn JP, Ghahremani S, Griggs R, Zaritsky J, et al. Contrast-enhanced magnetic resonance venography in pediatric patients with chronic kidney disease: initial experience with ferumoxytol. Pediatric radiology. 2016 doi: 10.1007/s00247-016-3605-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Uslu L, Donig J, Link M, Rosenberg J, Quon A, Daldrup-Link HE. Value of 18F-FDG PET and PET/CT for evaluation of pediatric malignancies. Journal of nuclear medicine : official publication, Society of Nuclear Medicine. 2015;56(2):274–86. doi: 10.2967/jnumed.114.146290. [DOI] [PubMed] [Google Scholar]
- 93.Marso SP, Hiatt WR. Peripheral arterial disease in patients with diabetes. Journal of the American College of Cardiology. 2006;47(5):921–9. doi: 10.1016/j.jacc.2005.09.065. [DOI] [PubMed] [Google Scholar]
- 94.Nathan DP, Tang GL. The impact of chronic renal insufficiency on vascular surgery patient outcomes. Seminars in vascular surgery. 2014;27(3–4):162–9. doi: 10.1053/j.semvascsurg.2015.01.006. [DOI] [PubMed] [Google Scholar]
- 95.Bashir MR, Jaffe TA, Brennan TV, Patel UD, Ellis MJ. Renal transplant imaging using magnetic resonance angiography with a nonnephrotoxic contrast agent. Transplantation. 2013;96(1):91–6. doi: 10.1097/TP.0b013e318295464c. [DOI] [PubMed] [Google Scholar]
- 96.Sigovan M, Gasper W, Alley HF, Owens CD, Saloner D. USPIO-enhanced MR angiography of arteriovenous fistulas in patients with renal failure. Radiology. 2012;265(2):584–90. doi: 10.1148/radiol.12112694. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Alam SR, Shah AS, Richards J, Lang NN, Barnes G, Joshi N, et al. Ultrasmall superparamagnetic particles of iron oxide in patients with acute myocardial infarction: early clinical experience. Circ Cardiovasc Imaging. 2012;5(5):559–65. doi: 10.1161/CIRCIMAGING.112.974907. [DOI] [PubMed] [Google Scholar]
- 98.Yilmaz A, Dengler MA, van der Kuip H, Yildiz H, Rosch S, Klumpp S, et al. Imaging of myocardial infarction using ultrasmall superparamagnetic iron oxide nanoparticles: a human study using a multi-parametric cardiovascular magnetic resonance imaging approach. European heart journal. 2013;34(6):462–75. doi: 10.1093/eurheartj/ehs366. [DOI] [PubMed] [Google Scholar]
- 99.Han F, Rapacchi S, Khan S, Ayad I, Salusky I, Gabriel S, et al. Four-dimensional, multiphase, steady-state imaging with contrast enhancement (MUSIC) in the heart: a feasibility study in children. Magnetic resonance in medicine. 2015;74(4):1042–9. doi: 10.1002/mrm.25491. [DOI] [PubMed] [Google Scholar]
- 100.Cheng JY, Hanneman K, Zhang T, Alley MT, Lai P, Tamir JI, et al. Comprehensive motion-compensated highly accelerated 4D flow MRI with ferumoxytol enhancement for pediatric congenital heart disease. Journal of magnetic resonance imaging : JMRI. 2016;43(6):1355–68. doi: 10.1002/jmri.25106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Hanneman K, Kino A, Cheng JY, Alley MT, Vasanawala SS. Assessment of the precision and reproducibility of ventricular volume, function, and mass measurements with ferumoxytol-enhanced 4D flow MRI. Journal of magnetic resonance imaging : JMRI. 2016 doi: 10.1002/jmri.25180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.DeSart K, O’Malley K, Schmit B, Lopez MC, Moldawer L, Baker H, et al. Systemic inflammation as a predictor of clinical outcomes after lower extremity angioplasty/stenting. J Vasc Surg. 2015;5(15):00954–4. doi: 10.1016/j.jvs.2015.04.399. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Okura H, Takagi T, Yoshida K. Therapies targeting inflammation after stent implantation. Curr Vasc Pharmacol. 2013;11(4):399–406. doi: 10.2174/1570161111311040004. [DOI] [PubMed] [Google Scholar]
- 104.Habets J, Zandvoort HJ, Reitsma JB, Bartels LW, Moll FL, Leiner T, et al. Magnetic resonance imaging is more sensitive than computed tomography angiography for the detection of endoleaks after endovascular abdominal aortic aneurysm repair: a systematic review. European journal of vascular and endovascular surgery : the official journal of the European Society for Vascular Surgery. 2013;45(4):340–50. doi: 10.1016/j.ejvs.2012.12.014. [DOI] [PubMed] [Google Scholar]
- 105.Ersoy H, Jacobs P, Kent CK, Prince MR. Blood pool MR angiography of aortic stent-graft endoleak. AJR American journal of roentgenology. 2004;182(5):1181–6. doi: 10.2214/ajr.182.5.1821181. [DOI] [PubMed] [Google Scholar]
- 106.Ichihashi S, Marugami N, Tanaka T, Iwakoshi S, Kurumatani N, Kitano S, et al. Preliminary experience with superparamagnetic iron oxide-enhanced dynamic magnetic resonance imaging and comparison with contrast-enhanced computed tomography in endoleak detection after endovascular aneurysm repair. J Vasc Surg. 2013;58(1):66–72. doi: 10.1016/j.jvs.2012.12.061. [DOI] [PubMed] [Google Scholar]
- 107.Corot C, Robert P, Idee JM, Port M. Recent advances in iron oxide nanocrystal technology for medical imaging. Advanced drug delivery reviews. 2006;58(14):1471–504. doi: 10.1016/j.addr.2006.09.013. [DOI] [PubMed] [Google Scholar]
- 108.Blockley NP, Jiang L, Gardener AG, Ludman CN, Francis ST, Gowland PA. Field strength dependence of R1 and R2* relaxivities of human whole blood to ProHance, Vasovist, and deoxyhemoglobin. Magnetic resonance in medicine. 2008;60(6):1313–20. doi: 10.1002/mrm.21792. [DOI] [PubMed] [Google Scholar]
- 109.Landry R, Jacobs PM, Davis R, Shenouda M, Bolton WK. Pharmacokinetic study of ferumoxytol: a new iron replacement therapy in normal subjects and hemodialysis patients. American journal of nephrology. 2005;25(4):400–10. doi: 10.1159/000087212. [DOI] [PubMed] [Google Scholar]
- 110.Neuwelt EA, Varallyay CG, Manninger S, Solymosi D, Haluska M, Hunt MA, et al. The potential of ferumoxytol nanoparticle magnetic resonance imaging, perfusion, and angiography in central nervous system malignancy: a pilot study. Neurosurgery. 2007;60(4):601–11. doi: 10.1227/01.NEU.0000255350.71700.37. discussion 11–2. [DOI] [PubMed] [Google Scholar]
- 111.Akeson P, Nordstrom CH, Holtas S. Time-dependency in brain lesion enhancement with gadodiamide injection. Acta radiologica (Stockholm, Sweden : 1987) 1997;38(1):19–24. doi: 10.1080/02841859709171236. [DOI] [PubMed] [Google Scholar]
- 112.Aghighi M, Pisani LJ, Sun Z, Klenk C, Madnawat H, Fineman SL, et al. Speeding up PET/MR for cancer staging of children and young adults. European radiology. 2016 doi: 10.1007/s00330-016-4332-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.

















