Sei sulla pagina 1di 27

See

discussions, stats, and author profiles for this publication at: https://www.researchgate.net/publication/274900253

Clinical applications of iron oxide nanoparticles


for magnetic resonance imaging of brain
tumors

Article in Nanomedicine · March 2015


DOI: 10.2217/nnm.14.203 · Source: PubMed

CITATIONS READS

25 777

6 authors, including:

Michael Iv Nicholas A. Telischak


Stanford University Stanford University
29 PUBLICATIONS 174 CITATIONS 26 PUBLICATIONS 122 CITATIONS

SEE PROFILE SEE PROFILE

Samantha J Holdsworth Heike Daldrup-Link


Stanford University Stanford University
52 PUBLICATIONS 597 CITATIONS 274 PUBLICATIONS 6,326 CITATIONS

SEE PROFILE SEE PROFILE

Some of the authors of this publication are also working on these related projects:

Pediatric Molecular Imaging View project

Amplified Magnetic Resonance Imaging (aMRI) View project

All content following this page was uploaded by Heike Daldrup-Link on 20 January 2016.

The user has requested enhancement of the downloaded file.


Review
For reprint orders, please contact: reprints@futuremedicine.com

Clinical applications of iron oxide


nanoparticles for magnetic resonance
imaging of brain tumors

Current neuroimaging provides detailed anatomic and functional evaluation of brain Michael Iv*,1, Nicholas
tumors, allowing for improved diagnostic and prognostic capabilities. Some challenges Telischak1, Dan Feng2,
persist even with today’s advanced imaging techniques, including accurate delineation Samantha J Holdsworth3,
of tumor margins and distinguishing treatment effects from residual or recurrent Kristen W Yeom2
& Heike E Daldrup-Link2
tumor. Ultrasmall superparamagnetic iron oxide nanoparticles are an emerging tool 1
Department of Radiology, Stanford
that can add clinically useful information due to their distinct physiochemical features University & Stanford University Medical
and biodistribution, while having a good safety profile. Nanoparticles can be used as Center, Stanford, CA 94305, USA
a platform for theranostic drugs, which have shown great promise for the treatment 2
Pediatric Radiology Section, Department
of CNS malignancies. This review will provide an overview of clinical ultrasmall of Radiology, Lucile Packard Children’s
Hospital, Stanford University, Stanford,
superparamagnetic iron oxides and how they can be applied to the diagnostic and
CA 94305, USA
therapeutic neuro-oncologic setting. 3
Department of Radiology, Lucas Center,
Stanford University, Stanford, CA 94305,
Keywords:  brain tumor • ferucarbotran • ferumoxtran • ferumoxytol • macrophage • MRI USA
• ultrasmall superparamagnetic iron oxide nanoparticles *Author for correspondence:
Tel.: +1 650 725 5384
Fax: +1 650 498 5374
Neuroimaging plays an important role in between treatment effects that arise from miv@ stanford.edu
patients with brain tumors. It provides for chemoradiation and true disease progres-
detailed anatomic evaluation of intracra- sion [11–17] .
nial neoplasms for surgical localization and This article provides an overview of current
intervention, and it allows for biological and clinical applications of ultrasmall superpara-
physiological characterization of these lesions magnetic iron oxide (USPIO) nanoparticles
to diagnose, prognosticate, assess treatment for use in MRI of brain tumors. We begin by
effects and monitor therapeutic responses. introducing the physiochemical, biodistribu-
Several MRI techniques have been employed tion and safety profiles of these agents and
to acquire such information including imag- then discuss their diagnostic capabilities in
ing with gadolinium-based contrast agents, detecting and characterizing tumor, imaging
perfusion-weighted imaging, diffusion- treatment effects and monitoring response to
weighted imaging and proton magnetic reso- therapy. Within this context, we review the
nance (MR) spectroscopy [1–10] . However, preclinical and clinical data present in the lit-
some challenges remain with current imag- erature. Finally, we highlight key therapeutic
ing that can impact survival and quality of uses of iron oxide nanoparticles in the man-
life in patients with brain cancer. These chal- agement of central nervous system neoplasms
lenges include the abilities to precisely delin- including delivery of drugs and monitoring
eate tumor margins for surgical resection (as stem cell therapies.
tumor cells can often be found outside areas
of enhancement or nonenhancing T2 signal Iron oxide nanoparticles
abnormality); to differentiate benign post- Iron oxide nanoparticles currently used in the
operative changes (such as blood products clinical arena are classified into two groups
and enhancement) from potential residual based on their mean hydrodynamic particle
part of
tumor following surgery; and to distinguish size: superparamagnetic iron oxide (SPIO)

10.2217/NNM.14.203 © 2015 Future Medicine Ltd Nanomedicine (Lond.) (2015) 10(6), 993–1018 ISSN 1743-5889 993
Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

and ultrasmall superparamagnetic iron oxide (USPIO) Biodistribution & metabolism


particles. SPIOs have a mean particle diameter greater After intravenous injection, USPIO nanoparticles cir-
than 50 nm, whereas USPIOs have a mean diameter of culate in the lumen of blood vessels (vascular phase),
less than 50 nm [18] . Due largely in part to their smaller transiently escaping sequestration and phagocyto-
size and longer blood half-life, USPIOs are better suited sis by macrophages of the reticuloendothelial system
for neuro-oncologic imaging [19] and are the focus of (RES), which is comprised of the liver, spleen, lymph
this review. USPIO particle design is similar between nodes and bone marrow (Figure 1) . This phenom-
available formulations but key differences exist in the enon leads to a prolonged blood half-life compared
nature of the coating, particle size and particle charge. with the larger SPIO. USPIOs do not extravasate
As a class, USPIOs are characterized by high longitudi- across intact vascular endothelia into the interstitial
nal (r1 = 1/T1) and transverse (r2 = 1/T2) relaxivities. space because of their relatively large size [24,26,33] .
These ‘T1- and T2-reducing’ agents can be used to rep- As such, USPIOs (particularly ferumoxytol due to
resent the degree of contrast agent uptake within a tis- its ability to be administered as a bolus) can serve as
sue [20–22] . Higher relaxivity agents allow for improved good vascular blood pool agents during the first-pass
quality of imaging and lesion depiction [22,23] . (arterial) and equilibrium (delayed) phases of imag-
There are currently three USPIOs that are under- ing (Figure 2)  [34–38] . In tumors or other tissues with
going clinical evaluation as a MR contrast agent: increased microvascular permeability, the nanopar-
ferumoxtran-10, ferumoxytol and ferucarbotran ticles gradually cross a disrupted blood–tissue barrier
C  [21,24–26] . Table 1 summarizes the physiochemical and enter the interstitium (‘interstitial phase’), where
features of these USPIOs, none of which is approved they exert T1 and T2 signal effects on MR [24,26,33] .
for therapeutic or diagnostic use in children. The positive T1 signal effect (increased signal inten-
Ferumoxtran-10 (Sinerem™; Guerbet, Paris, France sity on T1) is dependent on the quantity of protons
and Combidex™, AMI-227; Advanced Magnetics, and their degree of interaction with USPIOs [24,39–41] .
MA, USA) is a first-generation USPIO that has to be Therefore, T1-effects are more pronounced for inter-
administered as a slow infusion in order to avoid hypo- stitial compared with intracellular iron. On the other
tensive side effects. It has the longest blood half-life of hand, the negative T2 and T2* signal effect (decreased
the three USPIOs. The agent has been used in Phase signal intensity on T2- and T2*-weighted MR images)
I to Phase III clinical trials, primarily for lymph node is less dependent on iron oxide compartmentalization
imaging  [27–30] , but did not achieve US FDA approval (Figure 3)  [42] . While the exact route of transport of
due to inconclusive impact on patient management USPIOs across an altered blood–brain barrier has not
and was discontinued by the pharmaceutical industry. yet been fully elucidated, proposed mechanisms that
Recently, ferumoxtran-10 has been re-introduced to may help to explain increased vascular permeability in
clinical trials in Europe. tumors include presence of endothelial fenestrations
Ferumoxytol (Feraheme™, AMI-7228, Advanced and/or interendothelial junctions and increased trans-
Magnetics) is a second-generation USPIO with a car- port of particles via a single vesicle or chain of vesicles
boxymethyl dextran coating that allows for adminis- through endothelial cells [43–47] . Moreover, the rate
tration as a bolus without mast cell degranulation [31] . of USPIO leakage across the blood–brain barrier also
Of the three USPIOs, ferumoxytol has the fastest r1 depends on the histologic type of tumor; for example,
and r2 relaxivity, allowing for improved lesion detec- Beaumont  et al. observed that during a specific time
tion compared with the other agents. On 30 June frame, ferumoxtran-10 remained intravascular in a rat
2009, the US FDA approved ferumoxytol for the treat- C6 glioma model but extravasated in a RG2 glioma
ment of iron deficiency anemia in adults with chronic model [48] .
kidney disease [32] . More recently, the agent was also Several hours to days after USPIO administra-
approved for anemia treatment in Canada and several tion, nanoparticles are slowly phagocytosed by
countries in Europe. Thus, ferumoxytol has the advan- macrophages in the tumor interstitium (‘cellular
tage of being immediately and widely available for phase’)  [33] . Iron oxides in macrophages cause a pre-
clinical imaging of inflammation and tumors through dominant T2 and T2* effect but have a markedly
utilization as an ‘off label’ agent. diminished T1-effect compared with that of the
Ferucarbotran C (Supravist™, SHU 555 C, Scher- interstitial phase [39,49] . Some residual ‘free’ iron
ing AG) is a carboxy-dextran-coated USPIO and has oxide particles in the interstitium diffuse back into
the smallest hydrodynamic diameter of the three the blood pool once their interstitial concentration
USPIOs. This agent has been approved for clinical exceeds their blood concentration. Other ‘free’ iron
use in Europe, but was recently discontinued by the oxides may be retained in necrotic tumor areas result-
pharmaceutical industry. ing in a marked T1-effect, which may help to dif-

994 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

ferentiate these necrotic areas from intracellular iron

relaxivity
oxides [Golovko et al., Unpublished Data] .

(mM -1s-1)
Following phagocytosis, intracellular nanoparticles

60‡
83†
53†
in macrophages are slowly metabolized and cleared by

r1 relaxivity r2
the RES with biodegradation predominantly occur-
ring within lysosomes [33,50–53] . Elemental iron (ferri-

(mM -1s-1)
tin) contained within the nanoparticles is incorporated
into the body’s iron stores or is transferred to plasma

38†

24‡
23†
transferrin for incorporation into hemoglobin within
red blood cells. The dextran coat of ferumoxtran-10

half-life in half-life in
is cleaved by intracellular dextranases and excreted

humans

10–14 h
Blood
almost exclusively through the kidneys. The small

>24 h

6h
remainder is excreted in feces [21,54] . The carboxy-
methyl dextran coating of ferumoxytol is negligibly
degraded by dextranases when it is used to treat ane-

rodents

67 min

35 min
Blood
mia, and the amount of low molecular weight dex-

2–3 h
trans actually released from the coating and retained
in the kidneys is probably quite low although this has

Hydrodynamic
not yet been well defined [Claire Corot & Jean-Marc Idee,
Pers. Comm.] . Due to this overall slow metabolic process,

size (nm)
MR signal intensities of RES tissues gradually return

20–50

28–32
to baseline after several weeks [33] . There is currently Table 1. Physiochemical features of ultrasmall superparamagnetic iron oxide nanoparticles.†

20
no data available that reports how long USPIOs are
retained in tumors.
core (nm)
5.8–6.2

6.4–7.2
Size of
crystal

Safety

<5
Since the development of the first USPIO over two
decades ago, USPIOs have been tested and used as a
MR contrast agent in thousands of patients [50] . They
Carboxy-dextran

Carboxy-dextran
are generally well-tolerated when used in single dose.
Bernd  et al. examined 37 Phase I to Phase III clini-
Dextran T-10

cal trials for ferumoxtran-10 in 1777 adults. A total


Coating

of 23.2% of all patients had at least one adverse event,


86.3% of which were graded as mild to moderate in
severity. Common adverse events included back pain,
pruritus, headache and urticaria. About 2.6% of the
Ferumoxytol (Feraheme™; Advanced Magnetics) AMI-
Combidex™; Advanced Magnetics, MA, USA) AMI-227
Ferumoxtran-10 (Sinerem™; Guerbert, Paris, France;

patients developed severe adverse events, although only


Ferucarbotran C (Supravist™; Schering AG, Berlin,

0.42% of these events were considered to be treatment-


related (e.g., decreased oxygen saturation, chest pain,
dyspnea and skin rash). There were 12 deaths, but only
one was attributed to ferumoxtran-10 (anaphylactic
shock)  [55] . These findings are consistent with a pre-
vious Phase III trial in 152 adults, in which 28% of
all patients had one or more adverse effects [28] . Other
USPIOs have similar safety profiles. A Phase II clinical
trial showed that 22% of 63 subjects receiving feru-
Data taken from [21,24,25,26].

glose (Clariscan™, Amersham Health, UK) developed


Germany) SHU 555 C

mild-to-moderate adverse events, and no subject devel-


oped serious adverse effects [56] . Ferucarbotran C was
At 0.47T and 40°C.
At 0.47T and 39°C.

also well tolerated in all subjects in a Phase I clinical


trial [57,58] .
While most USPIOs have only been tested as con-
Name

7228

trast agents using a single dose, ferumoxytol showed


a satisfactory safety profile in adult patients with

future science group www.futuremedicine.com 995


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

Vascular phase Interstitial phase Cellular phase

Disrupted
blood–brain barrier

Tumor blood vessel

Normal blood vessel

Intact blood–brain barrier

Endothelium Tumor cell

Macrophage Iron oxide nanoparticle

Figure 1. Biodistribution of ultrasmall superparamagnetic iron oxide nanoparticles. Ultrasmall superparamagnetic iron oxides
(USPIOs) distribute and remain in the intravascular space early after injection (vascular phase). Tumors that cause increased
permeability allow for slow leakage of these particles into the interstitial space (interstitial phase). An intact blood–brain barrier
shields USPIOs from entering the interstitium of normal brain tissue. Over time, USPIOs are progressively phagocytosed by cellular
agents such as tumor-associated macrophages/microglia, astrocytes (not shown) and, to a lesser extent, primary tumor cells (cellular
phase). The blood–brain barrier is depicted in a simplified form for illustration purposes.

chronic kidney disease when used in a two-dose edema, headache, edema, vomiting, abdominal pain,
regime  [32] . An FDA report following its approval as chest pain, cough, pyrexia, back pain, muscle spasm
an iron supplement showed that out of 1726 subjects and dyspnea [32] .
recruited in three randomized clinical trials, 3.7% The rates of adverse reactions observed with USPIOs
of patients developed adverse events associated with are similar to but slightly higher than those seen with
hypersensitivity (e.g., rash, urticaria, pruritus, wheez- gadolinium-based contrast agents. For the latter, the
ing) and only 0.2% developed serious hypersensitiv- frequency of all acute adverse events after administra-
ity reactions (anaphylaxis). Although hypotension is tion ranges from less than 1 to 2.4%, the vast majority
another concerning side effect with iron oxide use, it of which are mild in severity [59–61] . Individuals with
only occurred in 1.9% of all subjects. It is more com- a history of allergy or asthma or previous reaction to
mon with rapid bolus injections and can be alleviated gadolinium had increased adverse reaction rates: 3.7
or avoided with slow injections. Other common but and 21.3%, respectively [60] . Severe anaphylactoid
less-severe adverse events (observed in greater than reactions, on the other hand, are extremely rare occur-
1% of subjects) included nausea, dizziness, peripheral ring in only 0.001–0.01% of cases [59] . In an accumu-

996 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

lated series of 20 million administered doses of a gado- Bridgeport, CT (a city and state in the USA) [63] and
linium chelate, there were 55 cases (<0.01%) of severe 12 cases per million inhabitants in Denmark have been
anaphylaxis and nine deaths that may have been drug- reported [64] .
related [61] . In addition, the administration of gadolin- As of today, various clinical trials have suggested
ium chelates in patients with severe renal impairment that USPIOs (ferumoxytol in particular) can be used
is associated with the development of nephrogenic sys- safely as contrast agents in adults. Of importance, iron
temic fibrosis, a widespread fibrotic tissue disorder that oxides are slowly metabolized in lysosomes within
is potentially fatal [62] . The occurrence rate of this dis- macrophages, predominantly Kupffer cells within the
order in patients with end stage renal disease exposed liver, and are not excreted via the kidneys [21,52,65] . Ele-
to gadolinium is not yet known, although incidences mental iron is deposited to the body’s iron reserve and
of 4.3 cases per 1000 patient-years in and around in the liver in the form of ferritin and/or hemosiderin.

A B

Figure 2. MRI of normal brain following ferumoxytol injection. High resolution (A) pre- and (B) immediate
post-ferumoxytol-enhanced T2*-weighted image of a healthy volunteer acquired with a 3D multi-echo
gradient-recalled echo imaging sequence at 3 Tesla. Note improved visualization of vessels on the (B) post-
ferumoxytol image due to presence of intraluminal iron oxide nanoparticles. (C) Ferumoxytol-enhanced MR
T1-weighted image shows excellent visualization of vessels in the brain confirming the utility of this ultrasmall
superparamagnetic iron oxide as a good blood pool agent.

future science group www.futuremedicine.com 997


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

T2 FSE T2* FGRE FSPGR


A B C
Pre-contrast

D E F
1 h Post-contrast

G H I
24 h Post-contrast

Figure 3. MRI of a mouse brain model implanted with glioblastoma multiforme. Precontrast (A) T2 FSE, (B) T2* fGRE and (C) T1 FSPGR
images show the tumor mass in the left brain. The mass shows (D & E) decreased T2 and T2* signal intensity and (F) increased T1
signal intensity 1 h after ferumoxytol injection. 24 h after administration, there is persistent and further (G & H) decreased T2 and T2*
signal intensity and (I) increased T1 signal intensity. Of note, the high concentration of iron uptake in the tumor causes such a marked
T2* shortening effect that such areas appear dark even on T1-weighted images (F & I).
FGRE: Fast gradient-recalled-echo; FSE: Fast spin echo; FSPGR: Fast spoiled gradient-recalled-echo.

Ferritin is also incorporated into hemoglobin begin- usually seen after administration of high amounts of
ning 1–2 days after injection. While the iron compo- dextran-40 or dextran-70 [66,67] . Additionally, there is a
nent of USPIOs is not excreted through the kidneys theoretical risk of acute renal failure with ferumoxytol
following its biodegradation, the leftover coating mate- due to iron-induced oxidative stress, which has been
rial of feruxmotran-10 is eliminated in the urine [21,54] . implicated as a cause of acute renal failure in patients
As previously mentioned, it is not yet known how the with chronic kidney disease who receive intravenous
coating material of feruxomytol is exactly degraded and sucrose  [68,69] . It is also important to note that the
eliminated although it is suspected that the quantita- blood half-life of ferumoxytol increases and the body
tive amount of dextrans produced and retained in the clearance decreases with increasing doses. Moreover,
kidneys is low [claire corot and jean-marc idee, ferumoxytol is not removed with dialysis [70] . Despite
pers. comm.]. This may be important because as with these potential safety risks, no ferumoxytol-induced
other colloids, dextrans may induce acute renal failure, acute nephrotoxicity has been found in clinical stud-

998 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

ies  [32,70] . Ferumoxytol is, therefore, relatively safe to study by Moore et al. that found a positive linear cor-
use in patients with renal insufficiency and is not asso- relation (r = 0.929) between USPIO internalization
ciated with any risk of nephrogenic systemic fibrosis as and tumor cell proliferation rate across multiple dif-
is the case with gadolinium chelates [71] . In contrast, ferent cell lines including C6 glioma, 9L gliosarcoma,
relatively little is known about the safety profile of U87 glioma and J774 sarcoma lines [78] . For a group
iron oxides in the pediatric population. With regard of malignant brain tumors comprised of oligoden-
to severe adverse effects including the reported death drogliomas, high-grade gliomas and glioblastomas,
associated with ferumoxtran-10 injection, evidence Neuwelt  et al. showed that the peak intensity of T1
has suggested that infusion dose and rate may play a enhancement occurs between 24 and 28 h after feru-
role  [55,72] . Future studies using a lower infusion rate moxytol injection followed by progressively decreas-
may result in improved safety profiles. ing T1 enhancement which persisted even after 72 h
post injection [31] , likely corresponding to nanopar-
Brain tumor detection & characterization ticle accumulation in the tumor interstitium followed
USPIOs are characterized by specific tumor micro- by TAM phagocytosis and intracellular compartmen-
vascular permeability and perfusion, accumulation in talization (Figure 5) . Studies utilizing other USPIOs
tumor interstitium and retention in tumor-associated such as ferumoxtran-10 and monocrystalline iron
macrophages (TAM). Many histopathologic studies oxide (MION) particles, a USPIO used in the pre-
following sampling or resection of brain tumors in clinical setting, have demonstrated similar results
humans have shown iron oxide particles in both the with the peak intensity of T1 enhancement occur-
tumor interstitium and in peritumoral reactive cells ring at 24 h with enhancement waning between 4 and
such as astrocytes, dendritic cells, microglia, TAM 7 days [51,73,76] ; in these studies, ferumoxtran-10 was
and, to a much lesser degree, in viable primary tumor evaluated in humans with predominantly high-grade
cells  [19,73–77] . Similar findings have also been dem- gliomas, and MION was used in rats injected with C6
onstrated in rodent studies [51,78–82] . Table 2 reviews glioma cells.
and summarizes many of these clinical and preclini- In a preclinical study of 13 rats implanted with
cal studies. Of significance is the fact that USPIOs C6 glioma cells, Zimmer et al. showed increased
can be used to detect and quantify TAM [26] , which T1 hyperintensity and T2 hypointensity within the
play an important role in promotion of tumor angio- brain tumors, which were most pronounced within
genesis, tumor progression and metastasis [83–86] . In smaller tumors, following injection of MION. His-
fact, clinical studies have shown a correlation between tologic evaluation showed iron deposits predomi-
TAM density and poor prognosis, particularly with nantly within the cytoplasm of tumor cells, allow-
regard to malignancy of the breast, cervix and blad- ing for sharp demarcation of the tumor and normal
der  [87] . While evidence for a significant correlation brain interface [51] . In a follow-up study using 22 rats
between the number of TAM and prognosis has been injected with thymidine kinase-positive 9L gliosar-
conflicting for human gliomas [88,89] , quantification coma cells, Zimmer et al. again showed increased
of TAM densities in malignant gliomas has shown MION uptake within the cytoplasm of tumor cells
to be potentially useful for monitoring response to and endothelial cells but not within normal brain
new TAM-modulating immunotherapies [26] . None- or non-MION injected controls. A small amount of
theless, USPIOs can be used as both a blood pool iron was also noted in the tumor interstitium and
agent during the ‘early’ vascular phase to probe tumor in reactive glial cells at the periphery of the tumor.
angiogenesis and increased perfusion associated with Potential additional MION uptake by TAM was not
aggressive tumors, and as a cellular imaging agent at a investigated in these studies. Overall, the pattern of
later phase [21,26,40] . uptake within these tumors was heterogeneous with
Following progressive phagocytosis of iron oxide the largest concentration located at the tumor-brain
particles by macrophages, intracellular iron causes a interface  [81] . Similar MR signal changes and histo-
negative signal effect on T2- and T2*-weighted imag- logic findings were found in a study by Moore et al.
ing, while no observable difference is seen in tissues of ten rats implanted with 9L gliosarcoma cells and
without macrophage infiltration (Figure 4)  [33,40,90] . injected with long-circulating dextran-coated iron
Malignant or aggressive tumors that more readily oxide (LCDIO) particles. The use of tumor cells that
disrupt the integrity of the blood–brain barrier and expressed the green-fluorescent protein marker, anti-
recruit phagocytic inflammatory cells to the tumor dextran antibodies to visualize the dextran coating of
microenvironment may therefore have a different LCDIO particles in tumoral tissue and radiolabeled
appearance than benign or nonaggressive tumors on LCDIO particles allowed for compartmental analy-
post-USPIO imaging [19] . This is corroborated by a sis of iron oxide accumulation and distribution. In

future science group www.futuremedicine.com 999


Table 2. Summary of clinical and preclinical studies using ultrasmall superparamagnetic iron oxide nanoparticles in brain tumor imaging.

1000
Study (year) Type of USPIO used Subjects Results Ref.
  study
Rainov et al. Preclinical MION 66 rats, some injected • Iron staining showed uptake in the periphery of tumor following MION injection  [80]
(1995) with 9L gliosarcoma without bradykinin infusion
cells • Significant increase of uptake in center of tumor following bradykinin infusion,
presumably due to disruption of brain-tumor barrier
• MRI in select rats showed distribution of iron throughout the tumor, primarily
along the periphery
Zimmer et al. Preclinical MION 13 rats implanted with • Glioma cells showed increased uptake of iron oxides, demonstrated as  [51]
(1995) C6 glioma cells hyperintensity on T1 and hypointensity on T2
• Signal characteristics at MR peaked at 12 h after administration of MION and
lasted up to 5 days
• Iron staining showed uptake in tumors (cytoplasm of tumor cells) after MION
administration

Nanomedicine (Lond.) (2015) 10(6)


Zimmer et al. Preclinical MION 22 rats injected with TK- • MION accumulates in tumor cells with uptake predominantly along the periphery  [81]
(1997) positive 9L gliosarcoma • Good correlation between histology and MRI to evaluate iron oxide uptake
cells • MR showed hypointensity on T2* images along the tumor periphery with more
heterogeneous center following MION injection
Dennie et al. Preclinical MION Rats implanted with C6 • Higher ΔR2*/ΔR2 ratio obtained with MRI in tumors relative to contralateral gray  [121]
matter suggesting a higher relative density of larger vessels in tumors; good
Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

(1998) glioma cells


correlation with predicted histological assessment
Enochs et al. Clinical Ferumoxtran-10 4 human patients with • USPIO enhancement in tumors on T1 images less than with gadolinium [73]
(1999) brain tumors (3 primary • Less consistent findings on T2* UPSIO images
gliomas, 1 metastatic • Tumor enhancement on T1 decreased more rapidly with gadolinium than with
melanoma) USPIO over time leading to faster blurring of tumor margin
• In 1 biopsy case, areas of enhancement on T1 with USPIO corresponded to tumor;
iron deposits seen in interstitium and viable tumor cells
Moore et al. Preclinical MION 10 rats implated with • Accumulation of long-circulating dextran-coated iron oxide nanoparticles seen in [78]
(2000) 9L-GFP glioma cells tumor cells and tumor-associated macrophages
Knauth et al. Preclinical MION 36 rats divided into • Surgically induced intracranial contrast seen on T1 in all rats who were imaged  [94]
(2001) three groups with gadolinium (group 1)
• No hyperintensities caused by MION in normal rat brains or brains with surgically
induced lesions (group 2)
• No surgically induced enhancement seen in brains of rats who were initially
implanted with C6 glioma cells, partially resected and then imaged with MION
(group 3)
ADC: Apparent diffusion coefficient; BCNU: 1,3-bis(2-chloroethyl)-1-nitrosourea; DDAH: dimethylarginine dimethylaminohydrolase; MION: Monocrystalline iron oxide nanoparticles; MR: Magnetic resonance;
rCBV: Relative cerebral blood volume; SPIO: Superparamagnetic iron oxide nanoparticles; TTP: Time to peak; USPIO: Ultrasmall superparamagnetic iron oxide nanoparticles; VSI: Vessel size index.

future science group


Table 2. Summary of clinical and preclinical studies using ultrasmall superparamagnetic iron oxide nanoparticles in brain tumor imaging (cont.).
Study (year) Type of USPIO used Subjects Results Ref.
  study
Varallyay et al. Clinical Ferumoxtran-10 20 human patients with • No change in T1 or T2 signal intensity in 3 patients after ferumoxide  [19]
(2002) and Ferumoxides various brain tumors administration

future science group


(SPIO) • Comparable but more variable enhancement on T1 with ferumoxtran than with
gadolinium
• All malignant tumors showed ferumoxtran enhancement
• Histology from 1 case showed distribution of ferumoxtran particles similar to MR
findings with iron uptake primarily by parenchymal cells with fibrillary processes
(astrocytes vs microglia) at tumor margin
Kostourou Preclinical NC100150 Rats implanted with C6 • Greater R2* and R2 rates and larger blood volume in glioma model  [125]
et al. (2003) glioma cells overexpressing DDAH, which metabolizes inhibitors of nitric oxide synthesis,
suggesting increased vascular development; similar VSI between this model and
wild-type glioma model
Neuwelt et al. Clinical Ferumoxtran-10 7 patients with brain • All tumors showed increased T1 and loss of signal on T2* following USPIO  [76]
(2004) tumors (6 primary administration; the effect was more pronounced on T1, where USPIO enhancing
gliomas, 1 metastatic lesions persisted for 2–5 days
unspecified) • Histochemistry staining showed iron uptake in reactive astrocytes and
macrophages (not tumor cells)
• Two patients showed enhancing lesions with USPIO not observed with
gadolinium
Tropres et al. Preclinical Ferumoxtran-10 12 rats implanted with • Null blood volume in areas of necrosis [105]
(2004) C6 glioma cells • In viable tumors, lower blood volume and higher VSI than in the contralateral
cortical area
• In the periphery, equal or higher blood volume and higher VSI than in the
contralateral cortical area
• Good correlation between VSI measurements obtained by MRI and histology
Hunt et al. Clinical Ferumoxtran-10 2 human patients with • Persistent T1 enhancement in tumoral tissue 24 h after USPIO administration as  [74]
(2005) primary gliomas detected by intraoperative MRI
• Histochemistry staining in one patient showed iron accumulation within
peritumoral reactive astrocytes (not tumor cells)
Manninger Clinical Ferumoxtran-10 5 human patients with • Variable results on T1, T2 and T2* with one patient showing increased size [92]
et al. (2005) primary central nervous and intensity of enhancing lesions on T1 and low-intensity on T2 and T2*
system lymphoma following USPIO injection compared with gadolinium, two patients showing
little difference between the two agents and two patients showing increased

www.futuremedicine.com
enhancement of lesions with gadolinium compared with USPIO
ADC: Apparent diffusion coefficient; BCNU: 1,3-bis(2-chloroethyl)-1-nitrosourea; DDAH: dimethylarginine dimethylaminohydrolase; MION: Monocrystalline iron oxide nanoparticles; MR: Magnetic resonance;
rCBV: Relative cerebral blood volume; SPIO: Superparamagnetic iron oxide nanoparticles; TTP: Time to peak; USPIO: Ultrasmall superparamagnetic iron oxide nanoparticles; VSI: Vessel size index.
Iron oxides for neuro-oncology 

1001
Review
Table 2. Summary of clinical and preclinical studies using ultrasmall superparamagnetic iron oxide nanoparticles in brain tumor imaging (cont.).

1002
Study (year) Type of USPIO used Subjects Results Ref.
  study
Muldoon et al. Preclinical Ferumoxtran-10, Rats • Evaluated imaging characteristics, distribution, time course and safety of USPIOs  [79]
(2005) Ferumoxytol, and one SPIO following intracerebral injection and transvascular delivery in rat
MION, brains
Ferumoxides • No parenchymal injury after iron oxide delivery
(SPIO) • Ferumoxtran showed best visualization of brain tumor in rats implanted with
LX-1 SCLC
compared with ferumoxytol and ferumoxides; iron staining showed uptake in
reactive cells at the tumor margin and within necrotic areas with ferumoxtran
and ferumoxytol
• Less enhancement seen with other tumor models (U87 glioblastoma and CALU6
SCLC) after ferumoxtran infusion; iron staining on U87 showed uptake along the
margin of tumor and iron staining on CALU6 showed minimal uptake

Nanomedicine (Lond.) (2015) 10(6)


Murillo et al. Clinical Ferumoxtran-10 55 human patients with • T2* images less informative than T1 due to iron-induced magnetic susceptibility  [75]
(2005) 56 intracranial lesions artifact
(various brain tumors) • Increased tumor enhancement on T1 with USPIO comparable to gadolinium (seen
in 19/34 glial tumors and 7/22 nonglial tumors)
• Most iron deposits were seen in inflammatory cells in cases with surgery
although some were seen in tumor cells
Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

Taschner et al. Clinical Ferumoxtran-10 9 human patients with • Heterogeneous and variable pattern of USPIO enhancement on T1 when [77]
(2005) various brain tumors compared with gadolinium
• Lack of USPIO enhancement on T1 in areas of presumed radiation necrosis
• Improved delineation of vascular structures on T2* USPIO images
• Histochemistry staining showed intracellular iron deposits within macrophages
(not in tumor) in 2 patients, iron-laden macrophages at the tumor-host interface
in 1 patient, few iron deposits in intratumoral macrophages in one patient and
absence of iron uptake in 5 patients
Claes et al. Preclinical Ferumoxtran-10 Mice implanted with • In untreated group, vessel leakage within tumor and relative high tumor  [100]
(2007) U87 glioma cells, blood volume yielded good visibility with gadolinium and T2* USPIO images,
split into treatment respectively
(vandetanib) and • Loss of tumor detectability with gadolinium in treated group due to restoration
nontreatment groups of blood–brain barrier following treatment, although decreased blood volume
could be detected with T2* USPIO
Kremer et al. Preclinical Ferumoxtran-10 40 mice implanted with • Increased enhancement on T1 along the margins of tumor following USPIO  [82]
(2007) four xenografts (TCG2, administration, corresponding to iron uptake within macrophages and microglia
TCG3, TCG4, U87) (not within tumor cells) on iron stains
ADC: Apparent diffusion coefficient; BCNU: 1,3-bis(2-chloroethyl)-1-nitrosourea; DDAH: dimethylarginine dimethylaminohydrolase; MION: Monocrystalline iron oxide nanoparticles; MR: Magnetic resonance;
rCBV: Relative cerebral blood volume; SPIO: Superparamagnetic iron oxide nanoparticles; TTP: Time to peak; USPIO: Ultrasmall superparamagnetic iron oxide nanoparticles; VSI: Vessel size index.

future science group


Table 2. Summary of clinical and preclinical studies using ultrasmall superparamagnetic iron oxide nanoparticles in brain tumor imaging (cont.).
Study (year) Type of USPIO used Subjects Results Ref.
  study
Neuwelt et al. Clinical Ferumoxytol 12 patients with • Maximal intensity of ferumoxytol enhancement was at 24–28 h following  [31]
(2007) malignant brain tumors administration

future science group


(10 with primary • Gadolinium leaks out of blood vessels early in contrast to ferumoxytol which
gliomas, 1 with breast remains intravascular in the ‘early’ phase after injection; therefore, ferumoxytol
cancer metastasis, 1 better evaluated tumor perfusion
with pineal tumor)
Quarles et al. Preclinical MION 18 rats implanted with • SU11657 group showed significantly decreased mean vessel diameter, decreased [126]
(2007) 9L gliosarcoma cells, blood volume, decreased mean transit time and increased blood flow
split into treatment
(SU11657) and control
groups
Gambarota Preclinical Ferumoxtran-10 Mice implanted with • All three tumor models showed increased delineation of tumor as well as [104]
et al. (2008) E34 glioblastoma evidence of vascular leakage and high blood volume following gadolinium and
xenograft, U87 glioma USPIO administration; confirmed with immunohistochemical stainings
cells and Mel57- • Mel57-VEGF-A165 showed the highest blood volume
VEGF-A165 (melanoma) • U87 showed higher blood volume in the rim than in the core
cells • Tumor vascular morphology best seen with T2* USPIO images
Serduc et al. Preclinical Ferumoxtran-10 70 mice implanted • Microbeam radiation therapy group showed significant increase in mean survival  [127]
(2008) with 9L gliosarcoma time and increase in ADC 24 h after treatment
cells, split into • Radiation therapy delays the time it takes for VSI to increase in treated as
treatment (microbream compared with untreated tumors
radiation therapy) and • No significant difference in blood volume between treated and untreated
nontreatment groups tumors
Beaumont Preclinical Ferumoxtran-10 14 rats implanted with • VSI MRI using USPIO can be combined with dynamic contrast-enhanced MRI  [48]
et al. (2009) C6 glioma cells and 6 using gadolinium to characterize tumor microvasculature
rats with RG2 glioma • Blood volume fractions and VSI of C6 and RG2 gliomas correlated well with
cells histology
• Good correlation of Ktrans value obtained with gadolinium ± USPIO for C6 glioma
but not for RG2 glioma (USPIO extravasation observed in the latter model)
Christoforidis Preclinical Ferucarbotran C 27 rats implanted with • Significantly increased visualization of tumor vascularity on T2* images with [103]
et al. (2009) F98 glioma cells USPIO, confirmed with histopathology
Varallyay et al. Preclinical Ferumoxytol 17 rats implanted with • Bevacizumab significantly decreased rCBV and increased TTP (indicating  [102]
(2009) U87MG human glioma decreased permeability) in tumors; rCBV assessed with dynamic MRI

www.futuremedicine.com
cells with ferumoxytol and vascular permeability assessed with dynamic
gadolinium-enhanced MRI
ADC: Apparent diffusion coefficient; BCNU: 1,3-bis(2-chloroethyl)-1-nitrosourea; DDAH: dimethylarginine dimethylaminohydrolase; MION: Monocrystalline iron oxide nanoparticles; MR: Magnetic resonance;
Iron oxides for neuro-oncology 

rCBV: Relative cerebral blood volume; SPIO: Superparamagnetic iron oxide nanoparticles; TTP: Time to peak; USPIO: Ultrasmall superparamagnetic iron oxide nanoparticles; VSI: Vessel size index.

1003
Review
1004
Table 2. Summary of clinical and preclinical studies using ultrasmall superparamagnetic iron oxide nanoparticles in brain tumor imaging (cont.).
Study (year) Type of USPIO used Subjects Results Ref.
  study
Bouchet et al. Preclinical Ferumoxtran-10 59 rats implanted with • Microbeam radiation therapy increased the median survival time of treated rats  [124]
(2010) 9L gliosarcoma cells and significantly increased blood vessel; decreased blood volume fraction and
vessel diameter in tumors were also observed from 8 days after treatment
• Preferential damage of tumor vessels vs normal vessels with microbeam radiation
therapy
Dosa et al. Clinical Ferumoxytol 26 human patients with • USPIO enhancement on T1 in all tumors at 24 h post injection; only 16 patients  [91]
(2011) various brain tumors showed T2 hypointensity
• Lesions seen in 6 patients with USPIO in areas without gadolinium enhancement
• Ferumoxytol-rCBV significantly higher compared with gadolinium-rCBV
Gahramanov Preclinical Ferumoxytol 13 rats implanted with • rCBV of vascularized tumors with disrupted brood brain barrier is  [101]
et al. (2011) U87MG tumor cells underestimated with gadolinium without the preload method and is dose
dependent with preload

Nanomedicine (Lond.) (2015) 10(6)


• rCBVs were high and independent of preload with ferumoxytol
• Decrease of rCBV after bevacizumab treatment was dependent on dose of
preload with gadolinium whereas it was constant and independent of preload
with ferumoxytol
Lemasson Preclinical Ferumoxtran-10 60 rats implanted • Higher ADC, blood volume fraction and VSI in tumor tissue than in contralateral  [112]
Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

et al. (2011) with U87MG human tissue


glioblastoma cells, • Sorafenib group showed decreased blood volume fraction, increased VSI,
randomized into increased ADC and decreased tumor growth rate 4–14 days after treatment
three equally sized • BCNU group showed increased ADC and decreased tumor growth 14 days after
groups (treatment treatment; no significant change in microvascular properties
with sorafenib,
treatment with BCNU
chemotherapy, no
treatment)
Pannetier Preclinical Ferumoxtran-10 12 rats implanted with • Comparable results of VSI in brain tumor tissues and contralateral tissues [123]
et al. (2012) C6 glioma cells obtained with dynamic susceptibility contrast MRI using gadolinium and steady-
state MRI using USPIO
• Higher VSI in brain tumor tissues than in contralateral tissue
Tani et al. Preclinical P904 9 rhesus monkeys • Successful localization of subthalamic nuclei with contrast enhanced MRI  [146]
(2011) using P904 (using subtraction images obtained from pre and post T2* images
following USPIO injection)
Burrell et al. Preclinical Ferumoxtran-10 24 rats implanted • Significant decrease in fractional blood volume, decrease in plasma perfusion, [147]
(2012) with C6 glioma cells increase in hemodynamic functionality (ΔR2*carbogen) and increase in tumor
(12 treated with hypoxia following treatment
cediranib and 12 treated
with vehicle)

future science group


ADC: Apparent diffusion coefficient; BCNU: 1,3-bis(2-chloroethyl)-1-nitrosourea; DDAH: dimethylarginine dimethylaminohydrolase; MION: Monocrystalline iron oxide nanoparticles; MR: Magnetic resonance;
rCBV: Relative cerebral blood volume; SPIO: Superparamagnetic iron oxide nanoparticles; TTP: Time to peak; USPIO: Ultrasmall superparamagnetic iron oxide nanoparticles; VSI: Vessel size index.
Table 2. Summary of clinical and preclinical studies using ultrasmall superparamagnetic iron oxide nanoparticles in brain tumor imaging (cont.).
Study (year) Type of USPIO used Subjects Results Ref.
  study
Thompson Clinical Ferumoxytol 7 human patients (4 • Use of ferumoxytol for dynamic susceptibility weighted contrast MR and  [99]
et al. (2012) with primary gliomas, 3 gadolinium for dynamic contrast-enhanced MR during a single imaging session is

future science group


with medulloblastomas) clinically useful to assess tumor perfusion and vascular permeability
• Tumor rCBV <1.75 suggestive of pseudoprogression rather than true progression
Gahramanov Clinical Ferumoxytol 19 human patients • With ferumoxytol and gadolinium with leakage correction, tumor rCBV values [97]
et al. (2013) with progressive ≤1.75 correlated with significantly improved survival; improved survival that was
glioblastoma following not statistically significantly was also seen with gadolinium without leakage
chemoradiation correction
Lemasson Preclinical Ferumoxtran-10 15 rats implanted with • Good correlation between steady state MRI and histology with regard to mean [122]
et al. (2013) C6 glioma cells and 12 vessel diameter, density and VSI
rats implanted with RG2 • Both tumor models showed increase in mean vessel diameter and VSI and
glioma cells decrease in density
Gutova M Preclinical Ferumoxytol Mice implanted with • T2-weighted MRI showed dynamic in vivo distribution of neural stem cells  [141]
et al. (2013) U251T.eGFP.ffluc human labeled with heparin, protamine sulfate and ferumoxytol at multiple time points
glioma cells following injection into glioma-bearing mice; results correlated with histology
• USPIO safe for clinical use
Varallyay et al. Clinical Ferumoxytol 49 human patients with • Consistent rCBV values in normal brain and tumor between steady-state imaging  [107]
(2013) various brain tumors with ferumoxytol (using a dose of 510 mg) and leakage-corrected dynamic
susceptibility contrast perfusion imaging with gadolinium; the former technique
also provided higher resolution images
ADC: Apparent diffusion coefficient; BCNU: 1,3-bis(2-chloroethyl)-1-nitrosourea; DDAH: dimethylarginine dimethylaminohydrolase; MION: Monocrystalline iron oxide nanoparticles; MR: Magnetic resonance;
rCBV: Relative cerebral blood volume; SPIO: Superparamagnetic iron oxide nanoparticles; TTP: Time to peak; USPIO: Ultrasmall superparamagnetic iron oxide nanoparticles; VSI: Vessel size index.

www.futuremedicine.com
Iron oxides for neuro-oncology 

1005
Review
Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

general, the USPIO was distributed throughout the lial cells (6.5%) [78] . Other studies revealed that iron
tumors, with the greatest amount occurring in the oxide uptake occurs chiefly in peritumoral and intra-
tumor periphery. Iron oxide nanoparticles were iden- tumoral macrophages/microglia and astrocytes with
tified in both the extracellular (comprised of the vas- minimal or no uptake in primary tumor cells [79,82] .
cular and interstitial spaces accounting for 23.5% of This suggests that areas of predominant T2 enhance-
the total tumor area) and intracellular (accounting ment and little T1 enhancement on delayed MR
for 76.5% of the total tumor area) compartments. images (>24h post nanoparticle injection) are
Intracellular compartmentalization occurred prefer- representative of TAM.
entially in glioma tumor cells (49%) and to a lesser With regard to the locoregional distribution of
degree in TAM (21%) and tumor vascular endothe- USPIO, Rainov et al. reported that the highest den-

Precontrast-T1w and T2w Gadoteridol 25-min ferumoxytol 24-h ferumoxytol

A B C D

E F G H

I rCBV – gadoteridol J rCBV – ferumoxytol High

Low

Figure 4. MRI characteristics of a patient with glioblastoma multiforme. (A) Precontrast and (B) gadoteridol-
enhanced T1-weighted images demonstrate an avidly enhancing left frontoparietal tumor crossing midline.
Ferumoxytol-enhanced T1-weighted image obtained (C) 25 min after injection shows faint areas of enhancement
and (D) 24 h after injection shows mixed signal intensity in the region where gadoteridol enhancement is seen. (E)
Precontrast and (F) gadoteridol-enhanced T2-weighted images. T2-weighted image obtained (G) 25 min and (H) 24 h
after ferumoxytol injection. Marked areas of decreased T2 signal are present 24 h after ferumoxytol injection. The
distribution of decreased T2 signal is similar to the area of mixed signal intensities on T1. (I & J) Color maps of relative
cerebral blood volume shows (J) high relative cerebral blood volume within the tumor on the postferumoxtyol image
but not on the (I) postgadoteridol image. The postgadoteridol image did not undergo leakage correction.
rCBV: Relative cerebral blood volume.
Reproduced with permission from [91] .

1006 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

A Diameters of ferumoxytol enhancement B C


(5 GBM patients average)
Relative average diameters

2.0
1.8
1.6 T2
Fe
1.4
1.2
1.0
0.8
Gd
0.6
0.4
0.2
0
0 20 40 60 80
Time (h)

D E F G H

Figure 5. Timeline of ferumoxytol enhancement in a patient with glioblastoma multiforme. (A) Line graph
comparing average diameter changes of ferumoxytol enhancement (solid line), average diameter changes of
gadolinium enhancement (dashed) and T2-weighted signal abnormalities (dash dot) over time. (B) Gadolinium-
enhanced T1-weighted, (C) T2-weighted and (D–H) ferumoxytol-enhanced T1-weighted images of a patient with
GBM. MRIs in (D–H) were obtained at the following time points: (D) 4–6 h, (E) 6–20 h, (F) 24–28 h, (G) 48–52 h
and (H) >72 h.
GBM: Glioblastoma multiforme.
Reproduced with permission from [31] .
sity of iron stained cells following MION admin- results when compared with the preclinical studies,
istration was at the periphery of glioma xenografts, in large part due to the heterogeneous population of
which correlated with the area of highest microvas- brain tumors included in the studies [19,73–77,91,92] .
cular density. Since vascularization in most human Dosa  et al. showed that T1 enhancement was seen
gliomas is different, animal models are needed that in all 26 patients with benign (n = 3, comprised of
better reflect human tumor physiology. However, two meningiomas and one pituitary adenoma) and
the authors did make an important observation: iron malignant (n = 23, comprised of 19 primary glio-
oxide nanoparticle delivery to the tumor center could mas, one angiocentric T-cell lymphoma and three
be significantly increased after bradykinin infusion, secondary metastasis) tumors 24 h post-ferumoxytol
which increases permeability of the brain tumor bar- injection, while T2 hypointensity was detected in
rier [80] . This intervention could be used for improved only 16 patients 24 h after administration [91] . The
delivery of theranostic nanoparticles to tumors with authors did note that they observed minimal to no
limited tumor microvascular permeability. signal intensity changes at the tumor margin of the
In comparison to animal and cell-culture mod- benign lesions although they did not further elabo-
els, clinical studies of human brain tumors evalu- rate. A possible explanation for the lack of T2 sig-
ated with USPIO-enhanced imaging also suggest nal seen in some of the masses may have been due
that USPIOs undergo intravascular and tumor to the relatively lower concentration of ferumoxytol
interstitium uptake as well as subsequent intracel- used (17 ml or 510 mg diluted with 17 ml of saline
lular compartmentalization. However, these stud- regardless of body weight), which may not have been
ies have some variation in imaging and histologic concentrated enough to produce distinct T2 signal

future science group www.futuremedicine.com 1007


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

effects. In addition, six patients had enhancement on Unlike malignant neoplasms, benign tumors tend
T1- and hypointensity on T2-weighted images 24 h to demonstrate minimal or no USPIO enhance-
post-ferumoxytol injection in areas around tumor ment  [91] . Varallyay et al. showed only slight to mod-
that did not enhance with gadolinium, perhaps due erate ferumoxtran enhancement of low-grade gliomas,
to intracellular accumulation of iron oxide particles hamartoma, meningioma and pituitary adenoma as
within macrophages. Following ferumoxtran-10 opposed to more prominent gadolinium enhance-
administration in five of seven patients with either ment observed with these lesions. This difference may
anaplastic oligodendroglioma or glioblastoma, Neu- be due to easier passage of smaller gadolinium mol-
welt et al. also demonstrated areas of T1 enhancement ecules through an incompetent blood–brain barrier as
on ferumoxtran-enhanced scans that were not pres- opposed to larger USPIO molecules as well as a rela-
ent on the gadolinium-enhanced scans; the enhance- tive paucity of phagocytic cells capable of accumulat-
ment extended beyond the margin of the main tumor ing the iron oxide particles [19] . Mixed results were
seen on the gadolinium images or was seen in new observed in a study of five patients with primary cen-
lesions altogether. In one of these patients with a mul- tral nervous system lymphoma who were given feru-
ticentric anaplastic oligodendroglioma, three tumor moxtran-10 [92] . Only one of the five subjects had more
foci that were identified on post-gadolinium images pronounced T1 and T2 signal changes post USPIO.
at five month follow-up had already been apparent In addition, Murillo et al. found variable enhance-
on the initial preoperative post-ferumoxtran images, ment in nonglial tumors with a tendency for more
but not on preoperative postgadolinium images [76] . aggressive tumors such as lymphoma, metastasis and
Extensive experience by this author and his group primitive neuroectodermal tumors to enhance more
has shown that tumor and its margin are best intensely after ferumoxtran-10 administration than
detected by increased signal on USPIO-enhanced with gadolinium (seen in five out of 22 patients) [75] .
T1-weighted images while T2-weighted images were This may again relate to the degree of blood–brain
less sensitive [19,75,76] . In addition, it was noted that barrier disruption. On the contrary, malignant glial
T2* images showed a ‘blooming effect’ (an apparent neoplasms are more infiltrative and proliferative; they
larger volume of tumor than seen on histology) due disrupt the blood–brain barrier, promote neoangio-
to magnetic susceptibility artifact induced by the genesis and recruit phagocytic inflammatory cells to
iron oxide nanoparticles [75] . the local environment, biological events that allow for
The appearance of brain metastasis often differs increased signal on USPIO-enhanced images.
in morphology from that of a malignant glioma on TAM in malignant gliomas have been associated
imaging. The degree of enhancement observed within with tumor aggressiveness and tumor grade [88] . The
a lesion depends on the histologic type of tumor vast majority of histochemistry studies performed
amongst other factors that can change the permeability in humans have overwhelmingly shown iron oxide
of the blood–brain barrier such as abnormal vascular- accumulation within peritumoral and intratumoral
ity or drug or radiation treatment [75] . Muldoon et al. reactive cells (macrophages/microglia and astro-
observed that postferumoxtran enhancement was dif- cytes), located primarily at the tumor margin, with
ferent in three intracerebral tumor models in rats (U87 minimal or no uptake within tumor cells [19,74–77] .
glioblastoma, LX-1 SCLC [small-cell lung carcinoma] Many of these studies only used cellular morphology
and CALU6 SCLC [a second SCLC tumor model]). at staining to determine specific cell types [19,74–76] .
Marked central ferumoxtran enhancement was seen One study [77] did use CD68, a marker that is highly
with the LX-1 SCLC tumor, which is a rapidly grow- expressed on human monocytes and tissue macro-
ing tumor, while minimal enhancement was seen phages  [93] , in two patients with malignant brain
with the other two models, which are slower growing tumors (anaplastic ependymoma and glioblastoma
tumors  [79] . Interestingly, the latter two models did multiforme) to show iron deposits located in TAM
show prominent gadolinium enhancement, suggest- while none was found in tumor cells nor in tumor
ing some differential leakiness of the blood–brain bar- interstitium. Therefore, ferumoxytol enhancement
rier or lack of inflammatory activity. Clinical studies in human tumors can serve as a biomarker for TAM.
have reported on the appearance of metastatic dis- Further studies in patients are needed to confirm evi-
ease in individual human patients following USPIO dence from preclinical studies that TAM enhance-
administration but the results have been variable with ment correlates with tumor aggressiveness and
enhancement observed in some lesions but not in oth- prognosis. One early study, however, did show iron
ers [31,73,75–77,91] . Future studies with larger sample size deposits within viable tumor cells and tumor intersti-
and histologic correlation are needed to fully investi- tium in a patient with recurrent grade III/IV mixed
gate the effect of USPIO in different metastatic tumors. cell glioma [73] , although it is unclear how tumor

1008 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

cells were identified or if there was any assessment the most widely used quantitative parameter derived
for the presence of TAM. from perfusion imaging for the assessment of tumor
The use of USPIOs in brain tumor imaging may vascular density and proliferation [1,95,96] . High rCBV
offer additional advantages over routine imaging with values are associated with increased tumor vascularity
gadolinium. First, due to its longer half-life, larger and viable tumor as well as more rapid time to progres-
size and decreased diffusion coefficient, USPIOs can sion of disease [7,95–97] . rCBV has been shown to be
cause persistent tumor enhancement over several days helpful in differentiating residual or recurrent tumor
thereby bypassing the need for additional contrast from treatment effects due to chemoradiation [97–99]
administration on short-term follow-up imaging. Gad- and can be used to monitor tumor response to anti­
olinium enhancement, on the other hand, peaks within angiogenic therapy [100–102] . Given these features,
minutes and resolves within hours leading to progres- rCBV may be utilized to determine short- and long-
sive blurring of tumor margins [73,74,76] . The prolonged term treatment strategies and to predict patient out-
ferumoxytol-tumor enhancement could be utilized to comes. Accurate measurement of tumor rCBV is,
guide surgeries with intraoperative MR exams. therefore, imperative.
The ability to accurately differentiate postsurgical USPIOs are good blood pool agents on the basis
changes following resection from residual tumor is of their long plasma half-life and macromolecular
yet another problem not entirely solved with today’s size, qualities that allow for longer circulation time in
standard imaging. In general, an area of gadolinium the intravascular space [21] . This allows for improved
enhancement that remains after resection of a tumor visualization of tumor vascularity and blood volume
in the early postoperative period (during the first four on MR [77,103–105] . In contrast to gadolinium, which
days after surgery) is worrisome for residual tumor [16] . is used in routine perfusion imaging, USPIOs do not
However, ‘benign’ surgically induced enhancement, cross a disrupted blood–brain barrier early after injec-
possibly a result of blood–brain barrier breakdown and tion (minutes to hours) [31,101] . This is an important
vascular injury, may also occur in the immediate post- quality for a reliable vascular contrast agent as early
operative period and complicates imaging interpreta- leakage may result in underestimation of rCBV [101] .
tion  [71,76] . In a preclinical study by Knauth et al., T1 Of the USPIOs, ferumoxytol has emerged as a good
enhancement with gadolinium was seen in all rat brains candidate for perfusion imaging because of its abil-
following the creation of surgically induced lesions. ity to be injected as a bolus [34] . Dosa et al. showed
However, after MION injection, no signal change on higher rCBV values obtained with ferumoxytol than
T1-weighted imaging was detected in a similar group. with gadolinium [91] . Comparable rCBV values were
This allowed for the detection of residual tumor using seen following ferumoxytol and gadolinium admin-
USPIO without the confounding presence of early istration but only after leakage correction techniques
postsurgical enhancement seen with gadolinium [94] . It were applied to the latter [97] . An alternative approach
is important to note, however, that some clinical stud- for obtaining CBV maps is steady-state susceptibility
ies have shown that the application of hemostatic and contrast imaging, which is feasible given ferumoxytol’s
oxidizing agents (such as Surgicel or H2O2) during sur- relatively long half-life and confinement to the intra-
gery can influence the signal of blood products in and vascular space early after injection. Unlike with DSC
around the resection cavity during the early postopera- imaging, the steady-state technique does not rely on
tive period, often with resulting prolonged T1 hyper- bolus tracking, bolus arrival, or transit times, thereby,
intense and T2 hypointense signal [16,17] . Residual avoiding the need to determine arterial input function.
USPIO enhancement may therefore be difficult to dif- Higher spatial resolution and distortion-free images
ferentiate from applied hemostatic agents and subacute can be obtained with the steady-state approach allow-
hemorrhage (which also appears T1 hyperintense) [76] , ing for improved characterization and localization of
and future studies are needed to address this potential malignant tumors associated with elevated cerebral
problem. blood volume [106,107] .
Residual or recurrent tumor may be differentiated
Imaging treatment effects & therapy from chemoradiation-induced changes with rCBV
monitoring measurements  [97–99] . Gahramanov et al. evaluated
Dynamic susceptibility-weighted contrast-enhanced patients with glioblastomas who underwent surgi-
(DSC) perfusion MRI is a helpful diagnostic tool in cal resection and chemoradiation and had conven-
brain tumor imaging and allows for the measurement tional MRI showing apparent tumor progression.
of tumor angiogenesis, a biomarker that can be used to With ferumoxytol, patients with areas of suspected
grade gliomas and assess the prognosis of patients with tumor showing low rCBV (≤1.75) had significantly
gliomas  [1] . Relative cerebral blood volume (rCBV) is improved survival than in those patients with tumors

future science group www.futuremedicine.com 1009


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

No contrast Gadoteridol

Before
CRT
0.1 1.0 3.0 5.0 7.0

Fe-rCBV Gd-rCBV Gd-rCBV LC

After
CRT

Fe Gd

Leakage map

Figure 6. Imaging of treatment effects (pseudoprogession) in a 73-year-old man with glioblastoma. Precontrast
and gadoteridol-enhanced T1-weighted magnetic resonance images obtained before and 3 months after
chemoradiotherapy demonstrate increased mass-like enhancement after treatment, concerning for disease
progression. The enhancing area shows no increase in rCBV on perfusion color maps obtained with ferumoxytol
(Fe-rCBV), gadoteridol (Gd-rCBV) and gadoteridol with leakage correction (Gd-rCBV LC) indicative of treatment
effects (pseudoprogression) rather than true disease progression. The leakage map shows gadoteridol contrast
leakage that can confound Gd-enhanced perfusion studies (arrow). No contrast leakage can be seen on the
ferumoxytol (Fe) image.
CRT: Chemoradiotherapy; rCBV: Relative cerebral blood volume.
Reproduced with permission from [97] .

showing high rCBV (>1.75). Low rCBV indicated progression (defined as rCBV >1.75). The authors
pseudoprogression (treatment effects), whereas found that the median survival was 34.7 months in
high rCBV indicated viable tumor (Figure 6) . Simi- patients with pseudoprogression and 13.4 months in
lar results were seen after gadolinium administra- patients without pseudoprogression. The longest sur-
tion. However, improved survival between the two vival (54.1 months) was seen in patients with both
rCBV groups became statistically significant with pseudoprogression and MGMT gene promoter meth-
gadolinium only after leakage correction techniques ylation [108] . The MGMT gene encodes a DNA-repair
were applied [97] . Thompson et al. also found that enzyme that is associated with resistance to alkylat-
low rCBV (<1) was suggestive of pseudoprogres- ing chemotherapeutic agents, and methylation of the
sion in a study of seven pediatric patients with brain promoter region of the gene is predictive of a more
tumors  [99] . In a recent retrospective observational favorable response to alkylating drugs in patients
study of 68 patients with newly diagnosed glioblas- with glioblastoma [109] . These results emphasize the
toma who were, for the most part, treated upfront importance of accurately differentiating pseudopro-
with combination temozolomide and radiation ther- gression from tumor progression and the significant
apy followed by adjuvant temozolomide, DSC perfu- role perfusion MRI may play in this assessment.
sion imaging with ferumoxytol was used to differ- Relative cerebral blood volume can also be used
entiate pseudoprogression (defined as rCBV <1.75 in to monitor response to antiangiogenic therapy,
an area of gadolinium enhancement) from tumor which has been used in many patients with high

1010 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

grade gliomas to target neovascularization. Among Therapeutic applications of this imaging technique
the antiangiogenic drugs, bevacizumab (Avastin™; in the preclinical setting have been used to evalu-
Genentech, CA, USA) is a monoclonal antibody that ate response to angiogenic stimulus (nitric oxide),
targets vascular endothelial growth factor [110] . Pre- antiangiogenic therapy, chemotherapy and radio-
clinical studies with ferumoxytol have shown reduc- therapy  [112,124–127] . Quarles et al. showed decreased
tion in rCBV values [101,102] and decreased permeabil- mean vessel diameter, blood volume and mean tran-
ity  [102] in tumors following bevacizumab treatment. sit time as well as increased blood flow in tumors
Decreased blood volume of tumors detected on T2*- treated with SU11657, an antiangiogenic drug [126] .
weighted imaging following ferumoxtran-10 admin- Lemasson  et al. observed that sorafenib, another
istration was also reported in mice implanted with antiangiogenic agent, increased VSI and decreased
U87 glioma cells and treated with vandetanib [100] . tumor growth rate 4–14 days after treatment, while
A Phase I clinical trial using ferumoxytol and gado- the cytotoxic drug, 1,3-bis(2-chloroethyl)-1-nitro-
linium to assess differences between the effects of sourea (BCNU), reduced tumor growth but had no
bevacizumab and dexamethasone in human subjects significant effect on microvascular properties [112] . In
with high-grade gliomas is ongoing [111] . Although two different studies, Serduc et al. and Bouchet et al.
the radiographic response of decreasing rCBV over showed that microbeam radiation therapy signifi-
time with antiangiogenic treatment may suggest cantly impacted the tumor microvasculature and
the successful targeting of angiogenesis, this effect increased the mean and median survival time of
has been shown to relate to normalization of tumor treated rats, respectively [124,127] . In human subjects,
blood vessels and may not hold any significance there is an overall paucity of data using vessel cali-
with respect to prognosis [112–115] . In early 2014, two ber imaging in the evaluation of brain tumors spe-
large Phase III multicenter randomized control tri- cifically with regard to imaging of treatment effects
als (AVAglio study and Radiation and RTOG 0825) (even less so with USPIOs), likely in part related to
evaluated the effect of adding bevacizumab to radio- the complexity of imaging acquisition which employs
therapy-temozolomide for the treatment of patients both gradient-echo and spin-echo techniques [119] .
with newly diagnosed glioblastoma and found no Although studies from Massachusetts General Hos-
improvement in overall survival in those patients pital have shown reduced vessel caliber after anti-
who received first-line bevacizumab therapy [116,117] . angiogenic therapy [113,119,128–130] , it is still unclear if
The AVAglio trial did find maintenance of baseline this technique will play an important role in predict-
quality of life and functional status and improved ing patient outcomes in this setting especially given
progression-free survival with bevacizumab use [116] . the results of the AVAglio and RTOG 0825 studies.
This is in contrast to the RTOG 0825 study, which Of note, it has been observed that influx of circulat-
reported a greater degree of neurocognitive function ing proangiogenic CD11b + monocytes into tumors
decline with bevacizumab therapy [117] . While there can restore depleted tumor vasculature by a process
are differing opinions for the current role of antian- known as ‘vasculogenesis’ [131–133] . USPIOs can visu-
giogenic drugs for the treatment of newly diagnosed alize this process and may help to guide improved or
glioblastomas, the trend appears to be to reserve alternative treatment strategies.
bevacizumab therapy for patients with recurrent
disease and/or for patients with moderate or severe Therapeutic uses of nanoparticles
neurologic symptoms [114,118] . USPIO nanoparticles are an emerging technology
Beyond rCBV mapping, USPIOs have been used to treat central nervous system malignancies. Their
to image the tumor microvasculature with MR-based capacity to be conjugated to antibodies and peptides
assessment of the mean vessel diameter, microvascu- allow for specific targeting of tumors and disruption
lar density and vessel size index (VSI) [119,120] . This is of associated active signaling pathways, delivery of
important as the formation and development of ves- drugs to target tissues across the blood–brain barrier,
sels in cancers play a significant role in their progres- monitoring and tracking neural stem cells used for
sion and in their response to antitumor therapy [119] . therapy and hyperthermic ablation of tumors in ther-
Several preclinical validation studies have shown motherapy. Peptides such as RGD and F3 have been
good correlation between MRI using iron oxide con- used to target nanoparticles to molecules on the sur-
trast agents and histologic determination of vessel face of glioma cells [134–136] . Larger peptides such as
caliber in C6 and RG2 glioma models [48,105,121,122] . monoclonal antibodies and cytokines have been used
Collectively, these studies have demonstrated as vehicles for drug delivery. Madhankumar et al.
increased VSI, which represents the average vessel effectively delivered the cytotoxic drug, doxorubicin,
caliber in an image voxel, in tumors [48,105,121–123] . to implanted gliomas in mice via liposomes conju-

future science group www.futuremedicine.com 1011


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

gated with human interleukin-13. Tumors in mice gadolinium chelates for patients with chronic kidney
receiving this method of treatment were found to disease. Future potential applications include cancer-
be smaller than in those injected with nontargeted targeted drug therapy and monitoring. The clinical
liposomes  [137] . Hadjipanayis et al. conjugated an utility of iron oxide nanoparticles is continuing to
antibody specific to the EGFRvIII deletion mutant evolve as we learn more about these powerful agents.
expressed by human glioblastoma cells to iron oxide
nanoparticles and delivered them to mice implanted Future perspective
with glioblastoma xenografts. Significant increase The future of USPIOs in neuro-oncology is excit-
in survival was found in animals after nanoparticle ing. Its use in diagnostic imaging may improve early
targeted cell therapy [138] . Neural stem cells have tumor detection, help distinguish between benign
additionally been used in conjunction with drugs and malignant tumors and further characterize
to target and treat gliomas and medulloblastomas tumor microenvironment. Future studies have to
in preclinical studies [139,140] . These stem cells can prove initial case reports that suggest that USPIOs
be monitored and tracked by preloading them with can delineate small malignant tumors that may not
USPIOs, a method which provides for easy identifi- be detected with conventional gadolinium-enhanced
cation and detection on susceptibility-sensitive T2- MRI. Differentiating infiltrating neoplasm from
and T2*-weighted MR sequences [140–142] . Another normal brain tissue is critical as this information
promising application of USPIOs is thermotherapy. could alter surgical options, including biopsies and
Hyperthermic ablation of tumors involves uptake resections. In addition, the potential of USPIOs to
of nanoparticles within a tumor with subsequent monitor therapeutic efficacy via accurate measure-
exposure to an alternating magnetic field, which ments of tumor perfusion is paramount in develop-
produces electrical current resulting in heat genera- ing and fine-tuning treatment strategies. Quantify-
tion [143,144] . A prospective, two-center Phase II clini- ing TAM through ferumoxytol-enhanced MRI can
cal trial found that a combination of thermotherapy be used to grade gliomas, predict prognosis and out-
using magnetic nanoparticles and reduced fraction- comes and guide immune-modulating cancer thera-
ated stereotactic radiotherapy in patients with recur- pies. The use of USPIOs in the treatment of central
rent glioblastoma resulted in longer overall survival nervous system neoplasms is especially provocative
following diagnosis of the first tumor recurrence. A as they can be used as vehicles for cancer-targeted
high concentration of nanoparticles (112 mg/ml) was chemotherapeutic drug delivery. Specifically target-
needed to generate sufficient heat within the tumor ing tumor cells and TAM with delivery of activatable
for effective thermotherapy, with a median peak cytotoxic drugs to these targets across a formidable
temperature of 51.2°C within the tumor [145] . blood–brain barrier can change the way we currently
treat malignancy. Other useful applications includ-
Conclusion ing monitoring and tracking stem cells and other
Ultrasmall superparamagnetic iron oxide nanopar- less conventional therapies offer different avenues in
ticles are an important tool for detecting and char- neuro-oncology.
acterizing brain tumors, imaging treatment effects
and monitoring treatment efficacy. Due to their Acknowledgements
relatively long circulation time, they can be used to The authors would like to thank Amy (Morris) Thomas for
better evaluate tumor vascularity without requiring helping them to design Figure 1, Michael Moseley, for provid-
repeat injections on short-term follow-up imaging. ing the images for Figure 2 and Olga Lenkov for providing the
Their capacity for early retention in the intravascu- images for Figure 3. They also thank Edward Neuwelt for all of
lar space also allows for more accurate assessment of his valuable suggestions and input.
tumor perfusion and determination of relative cere-
bral blood volume, a robust imaging marker that is Financial & competing interests disclosure
important in grading cancers and predicting patient The authors acknowledge and thank the Stanford Depart-
outcomes. Due to their phagocytosis by macro- ment of Radiology Angel Grant for generously supporting
phages, USPIOs can be used to quantify the intrin- their initial investigations. The authors have no other relevant
sic immune response to malignant gliomas, which is affiliations or financial involvement with any organization or
associated with tumor aggressiveness and prognosis, entity with a financial interest in or financial conflict with the
and to monitor new cancer immunotherapies. Thus subject matter or materials discussed in the manuscript apart
far, ferumoxytol has been well tolerated in patients, from those disclosed.
and its relative kidney-independent pharmacokinet- No writing assistance was utilized in the production of this
ics makes it a suitable alternative contrast agent to manuscript.

1012 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

Executive summary
Iron oxide nanoparticles
• Ultrasmall superparamagnetic iron oxide (USPIO) particles have a mean diameter of less than 50 nm and a
relatively long blood half-life.
• USPIOs are characterized by high longitudinal and transverse relaxivities, allowing for improved quality of
imaging and lesion depiction on MRI.
Biodistribution & metabolism
• USPIOs remain in the intravascular space early after injection, allowing it to serve as a good blood pool agent.
• They gradually cross a disrupted blood–brain barrier in tumors and into the interstitial space. They are
progressively phagocytosed by tumor-associated macrophages/microglia, astrocytes and, to a lesser extent,
tumor cells.
Safety
• USPIOs are well-tolerated by patients and have a good safety profile.
• USPIOs are eliminated by the reticuloendothelial system, predominantly by hepatic macrophages (Kupffer
cells). They do not get eliminated by the kidneys and are therefore deemed safe for use in adult patients with
chronic renal failure.
• Ferumoxytol is currently US FDA approved for the treatment of iron deficiency anemia in adults with chronic
kidney disease and is, therefore, widely available for use as an ‘off-label’ contrast agent.
Brain tumor detection & characterization
• Malignant gliomas show increased T1 signal intensity and decreased T2/T2* signal intensity on
postferumoxytol magnetic resonance images.
• Signal intensity on T1 peaks at approximately 24 h and persists for days in contrast to gadolinium-based
contrast agents.
Imaging treatment effects & therapy monitoring
• Ferumoxytol allows for accurate measurement of relative cerebral blood volume, an important imaging
marker used to grade gliomas and monitor treatment effects.
• Beyond cerebral blood volume mapping, USPIOs can be used to image the tumor microvasculature with MR-
determined values of mean vessel diameter, microvascular density and vessel size index.
Therapeutic uses of nanoparticles
• Therapeutic applications of USPIOs include direct targeting of tumors, delivery of drugs to tumors across
the formidable blood–brain barrier, monitoring and tracking neural stem cells and hyperthermic ablation of
tumors.

References 7 Law M, Young RJ, Babb JS et al. Gliomas: predicting


Papers of special note have been highlighted as: time to progression or survival with cerebral blood volume
• of interest; •• of considerable interest measurements at dynamic susceptibility-weighted contrast-
enhanced perfusion MR imaging. Radiology 247(2),
1 Cha S. Update on brain tumor imaging: from anatomy to
490–498 (2008).
physiology. Am. J. Neuroradiol. 27(3), 475–487 (2006).
8 Bulakbasi N, Kocaoglu M, Farzaliyev A, Tayfun C, Ucoz T,
• Provides excellent review on current state of brain tumor
Somuncu I. Assessment of diagnostic accuracy of perfusion
imaging.
MR imaging in primary and metastatic solitary malignant
2 Thakor AS, Gambhir SS. Nanooncology: the future of brain tumors. Am. J. Neuroradiol. 26(9), 2187–2199 (2005).
cancer diagnosis and therapy. CA Cancer J. Clin. 63(6),
9 Roberts HC, Roberts TP, Brasch RC, Dillon WP.
395–418 (2013).
Quantitative measurement of microvascular permeability
3 Provenzale JM, Silva GA. Uses of nanoparticles for central in human brain tumors achieved using dynamic contrast-
nervous system imaging and therapy. Am. J. Neuroradiol. enhanced MR imaging: correlation with histologic grade.
30(7), 1293–1301 (2009). Am. J. Neuroradiol. 21(5), 891–899 (2000).
4 Meyers JD, Doane T, Burda C, Basilion JP. Nanoparticles 10 Astrakas LG, Zurakowski D, Tzika AA et al. Noninvasive
for imaging and treating brain cancer. Nanomedicine 8(1), magnetic resonance spectroscopic imaging biomarkers to
123–143 (2013). predict the clinical grade of pediatric brain tumors. Clin.
5 Kono K, Inoue Y, Nakayama K et al. The role of diffusion- Cancer Res. 10(24), 8220–8228 (2004).
weighted imaging in patients with brain tumors. Am. 11 Cheng Y, Morshed RA, Auffinger B, Tobias AL, Lesniak MS.
J. Neuroradiol. 22(6), 1081–1088 (2001). Multifunctional nanoparticles for brain tumor imaging and
6 Yu CS, Li KC, Xuan Y, Ji XM, Qin W. Diffusion tensor therapy. Adv. Drug Deliv. Rev. 66, 42–57 (2014).
tractography in patients with cerebral tumors: a helpful 12 Covarrubias DJ, Rosen BR, Lev MH. Dynamic magnetic
technique for neurosurgical planning and postoperative resonance perfusion imaging of brain tumors. Oncologist
assessment. Eur. J. Radiol. 56(2), 197–204 (2005). 9(5), 528–537 (2004).

future science group www.futuremedicine.com 1013


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

13 Earnest FT, Kelly PJ, Scheithauer BW et al. Cerebral prostate cancer. N. Engl. J. Med. 348(25), 2491–2499
astrocytomas: histopathologic correlation of MR and CT (2003).
contrast enhancement with stereotactic biopsy. Radiology 28 Anzai Y, Piccoli CW, Outwater EK et al. Evaluation of neck
166(3), 823–827 (1988). and body metastases to nodes with ferumoxtran 10-enhanced
14 Nie G, Hah HJ, Kim G et al. Hydrogel nanoparticles with MR imaging: phase III safety and efficacy study. Radiology
covalently linked coomassie blue for brain tumor delineation 228(3), 777–788 (2003).
visible to the surgeon. Small 8(6), 884–891 (2012). 29 Heesakkers RA, Futterer JJ, Hovels AM et al. Prostate cancer
15 Spickler EM, Dion JE, Lufkin RB et al. The MR appearance evaluated with ferumoxtran-10-enhanced T2*-weighted MR
of endovascular embolic agents in vitro with clinical Imaging at 1.5 and 3.0 T: early experience. Radiology 239(2),
correlation. Comp. Med. Imaging Graph. 14(6), 415–423 481–487 (2006).
(1990). 30 Heesakkers RA, Hovels AM, Jager GJ et al. MRI with a
16 Forsting M, Albert FK, Kunze S, Adams HP, Zenner lymph-node-specific contrast agent as an alternative to CT
D, Sartor K. Extirpation of glioblastomas: MR and CT scan and lymph-node dissection in patients with prostate
follow-up of residual tumor and regrowth patterns. Am. cancer: a prospective multicohort study. Lancet Oncol. 9(9),
J. Neuroradiol. 14(1), 77–87 (1993). 850–856 (2008).
17 Spiller M, Tenner MS, Couldwell WT. Effect of absorbable 31 Neuwelt EA, Varallyay CG, Manninger S et al. The potential
topical hemostatic agents on the relaxation time of blood: an of ferumoxytol nanoparticle magnetic resonance imaging,
in vitro study with implications for postoperative magnetic perfusion, and angiography in central nervous system
resonance imaging. J. Neurosurg. 95(4), 687–693 (2001). malignancy: a pilot study. Neurosurgery 60(4), 601–611;
18 Benderbous S, Corot C, Jacobs P, Bonnemain B. discussion 611–602 (2007).
Superparamagnetic agents: physicochemical characteristics • Provides timeline for ferumoxytol enhancement in
and preclinical imaging evaluation. Acad. Radiol. malignant tumors observed on MRI.
3(Suppl. 2), S292–S294 (1996).
32 Lu M, Cohen MH, Rieves D, Pazdur R. FDA report:
19 Varallyay P, Nesbit G, Muldoon LL et al. Comparison Ferumoxytol for intravenous iron therapy in adult patients
of two superparamagnetic viral-sized iron oxide particles with chronic kidney disease. Am. J. Hematol. 85(5), 315–319
ferumoxides and ferumoxtran-10 with a gadolinium chelate (2010).
in imaging intracranial tumors. Am. J. Neuroradiol. 23(4),
•• Discusses important US FDA report reviewing the safety
510–519 (2002).
profile of ferumoxytol.
20 Beckmann N, Cannet C, Babin AL et al. In vivo visualization
33 Daldrup-Link H, Coussens LM. MR imaging of tumor-
of macrophage infiltration and activity in inflammation
associated macrophages. Oncol. Immunol. 1(4), 507–509
using magnetic resonance imaging. Wiley Interdiscip. Rev.
(2012).
Nanomed. Nanobiotechnol. 1(3), 272–298 (2009).
34 Stabi KL, Bendz LM. Ferumoxytol use as an intravenous
21 Corot C, Robert P, Idee JM, Port M. Recent advances in
contrast agent for magnetic resonance angiography. Ann.
iron oxide nanocrystal technology for medical imaging. Adv.
Pharmacother. 45(12), 1571–1575 (2011).
Drug Deliv. Rev. 58(14), 1471–1504 (2006).
35 Li W, Tutton S, Vu AT et al. First-pass contrast-enhanced
•• Provides excellent review of iron oxide nanoparticles.
magnetic resonance angiography in humans using
22 Giesel FL, Mehndiratta A, Essig M. High-relaxivity contrast- ferumoxytol, a novel ultrasmall superparamagnetic iron
enhanced magnetic resonance neuroimaging: a review. Eur. oxide (USPIO)-based blood pool agent. J. Magn. Reson.
Radiol. 20(10), 2461–2474 (2010). Imaging 21(1), 46–52 (2005).
23 Cotton F, Hermier M. The advantage of high relaxivity 36 Prince MR, Zhang HL, Chabra SG, Jacobs P, Wang Y. A
contrast agents in brain perfusion. Eur. Radiol. 16(Suppl. 7), pilot investigation of new superparamagnetic iron oxide
M16–M26 (2006). (ferumoxytol) as a contrast agent for cardiovascular MRI.
24 Simon GH, Von Vopelius-Feldt J, Fu Y et al. Ultrasmall J. Xray Sci. Technol. 11(4), 231–240 (2003).
supraparamagnetic iron oxide-enhanced Magn. Reson. 37 Ersoy H, Jacobs P, Kent CK, Prince MR. Blood pool MR
Imaging of antigen-induced arthritis: a comparative study angiography of aortic stent-graft endoleak. Am. J. Roentgenol.
between SHU 555 C, ferumoxtran-10, and ferumoxytol. 182(5), 1181–1186 (2004).
Invest. Radiol. 41(1), 45–51 (2006).
38 Li W, Salanitri J, Tutton S et al. Lower extremity deep
25 Jung CW, Jacobs P. Physical and chemical properties venous thrombosis: evaluation with ferumoxytol-enhanced
of superparamagnetic iron oxide MR contrast agents: MR imaging and dual-contrast mechanism – preliminary
ferumoxides, ferumoxtran, ferumoxsil. Magn. Reson. Imaging experience. Radiology 242(3), 873–881 (2007).
13(5), 661–674 (1995).
39 Simon GH, Bauer J, Saborovski O et al. T1 and T2 relaxivity
26 Daldrup-Link HE, Golovko D, Ruffell B et al. MRI of of intracellular and extracellular USPIO at 1.5T and 3T
tumor-associated macrophages with clinically applicable iron clinical MR scanning. Eur. Radiol. 16(3), 738–745 (2006).
oxide nanoparticles. Clin. Cancer Res. 17(17), 5695–5704
40 Corot C, Petry KG, Trivedi R et al. Macrophage imaging in
(2011).
central nervous system and in carotid atherosclerotic plaque
27 Harisinghani MG, Barentsz J, Hahn PF et al. Noninvasive using ultrasmall superparamagnetic iron oxide in magnetic
detection of clinically occult lymph-node metastases in resonance imaging. Invest. Radiol. 39(10), 619–625 (2004).

1014 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

41 Chambon C, Clement O, Le Blanche A, Schouman-Claeys oxide contrast agent: comprehensive analysis of a clinical
E, Frija G. Superparamagnetic iron oxides as positive MR development program. Invest. Radiol. 44(6), 336–342
contrast agents: in vitro and in vivo evidence. Magn. Reson. (2009).
Imaging 11(4), 509–519 (1993). 56 Daldrup-Link HE, Rydland J, Helbich TH et al.
42 Girard OM, Ramirez R, Mccarty S, Mattrey RF. Toward Quantification of breast tumor microvascular permeability
absolute quantification of iron oxide nanoparticles as well with feruglose-enhanced MR imaging: initial phase II
as cell internalized fraction using multiparametric MRI. multicenter trial. Radiology 229(3), 885–892 (2003).
Contrast Media Mol. Imaging 7(4), 411–417 (2012). 57 Reimer P, Bremer C, Allkemper T et al. Myocardial
43 Feng D, Nagy JA, Hipp J, Dvorak HF, Dvorak AM. perfusion and MR angiography of chest with SH U 555 C:
Vesiculo-vacuolar organelles and the regulation of venule results of placebo-controlled clinical phase i study. Radiology
permeability to macromolecules by vascular permeability 231(2), 474–481 (2004).
factor, histamine, and serotonin. J. Exp. Med. 183(5), 58 Tombach B, Reimer P, Bremer C et al. First-pass and
1981–1986 (1996). equilibrium-MRA of the aortoiliac region with a
44 Stewart PA, Hayakawa K, Hayakawa E, Farrell CL, Del superparamagnetic iron oxide blood pool MR contrast agent
Maestro RF. A quantitative study of blood-brain barrier (SH U 555 C): results of a human pilot study. NMR Biomed.
permeability ultrastructure in a new rat glioma model. Acta 17(7), 500–506 (2004).
Neuropathol. 67(1–2), 96–102 (1985). 59 Murphy KP, Szopinski KT, Cohan RH, Mermillod B, Ellis
45 Black KL, Ningaraj NS. Modulation of brain tumor JH. Occurrence of adverse reactions to gadolinium-based
capillaries for enhanced drug delivery selectively to brain contrast material and management of patients at increased
tumor. Cancer Control 11(3), 165–173 (2004). risk: a survey of the American Society of Neuroradiology
46 Renkin EM. Multiple pathways of capillary permeability. Fellowship Directors. Acad. Radiol. 6(11), 656–664 (1999).
Circ. Res. 41(6), 735–743 (1977). 60 Nelson KL, Gifford LM, Lauber-Huber C, Gross CA, Lasser
47 Schlageter KE, Molnar P, Lapin GD, Groothuis DR. TA. Clinical safety of gadopentetate dimeglumine. Radiology
Microvessel organization and structure in experimental brain 196(2), 439–443 (1995).
tumors: microvessel populations with distinctive structural 61 Niendorf H, Alhassan A, Balzer T, Claub W, Greens V.
and functional properties. Microvasc. Res. 58(3), 312–328 Safety and risk of gadolinium-DTPA: extended clinical
(1999). experience after more than 20 million applications. In:
48 Beaumont M, Lemasson B, Farion R, Segebarth C, Remy C, Magnevist.Felix R, Hosten N, Hricak H (Eds). Blackwell
Barbier EL. Characterization of tumor angiogenesis in rat Science, Berlin, Germany, 17–27 (1998).
brain using iron-based vessel size index MRI in combination 62 Kuo PH, Kanal E, Abu-Alfa AK, Cowper SE. Gadolinium-
with gadolinium-based dynamic contrast-enhanced MRI. based MR contrast agents and nephrogenic systemic fibrosis.
J. Cereb. Blood Flow Metabol. 29(10), 1714–1726 (2009). Radiology 242(3), 647–649 (2007).
49 Henning TD, Wendland MF, Golovko D et al. Relaxation 63 Deo A, Fogel M, Cowper SE. Nephrogenic systemic fibrosis:
effects of ferucarbotran-labeled mesenchymal stem cells at a population study examining the relationship of disease
1.5T and 3T: discrimination of viable from lysed cells. Magn. development to gadolinium exposure. Clin. J. Am. Soc.
Reson. Med. 62(2), 325–332 (2009). Nephrol. 2(2), 264–267 (2007).
50 Weissleder R, Elizondo G, Wittenberg J, Rabito CA, Bengele 64 Elmholdt TR, Olesen AB, Jorgensen B et al. Nephrogenic
HH, Josephson L. Ultrasmall superparamagnetic iron oxide: systemic fibrosis in Denmark‐‐a nationwide investigation.
characterization of a new class of contrast agents for MR PLoS ONE 8(12), e82037 (2013).
imaging. Radiology 175(2), 489–493 (1990). 65 Kalber TL, Smith CJ, Howe FA et al. A longitudinal study
51 Zimmer C, Weissleder R, Poss K, Bogdanova A, Wright SC of R2* and R2 Magnetic Resonance Imaging relaxation rate
Jr, Enochs WS. MR imaging of phagocytosis in experimental measurements in murine liver after a single administration of
gliomas. Radiology 197(2), 533–538 (1995). 3 different iron oxide-based contrast agents. Invest. Radiol.
52 Schulze E, Ferrucci JT Jr, Poss K, Lapointe L, Bogdanova 40(12), 784–791 (2005).
A, Weissleder R. Cellular uptake and trafficking of a 66 Biesenbach G, Kaiser W, Zazgornik J. Incidence of acute
prototypical magnetic iron oxide label in vitro. Invest. Radiol. oligoanuric renal failure in dextran 40 treated patients with
30(10), 604–610 (1995). acute ischemic stroke stage III or IV. Renal Fail. 19(1), 69–75
53 Levy M, Luciani N, Alloyeau D et al. Long term in vivo (1997).
biotransformation of iron oxide nanoparticles. Biomaterials 67 Schinco MA, Hughes D, Santora TA. Complications of 32%
32(16), 3988–3999 (2011). dextran-70 in 10% dextrose. A case report. J. Reprod. Med.
54 Bourrinet P, Bengele HH, Bonnemain B et al. Preclinical 41(6), 455–458 (1996).
safety and pharmacokinetic profile of ferumoxtran-10, an 68 Bishu K, Agarwal R. Acute injury with intravenous iron
ultrasmall superparamagnetic iron oxide magnetic resonance and concerns regarding long-term safety. Clin. J. Am. Soc.
contrast agent. Invest. Radiol. 41(3), 313–324 (2006). Nephrol. 1(Suppl. 1), S19–S23 (2006).
• Provides excellent review of ferumoxtran-10. 69 Pai AB, Garba AO. Ferumoxytol: a silver lining in the
55 Bernd H, De Kerviler E, Gaillard S, Bonnemain B. Safety treatment of anemia of chronic kidney disease or another
and tolerability of ultrasmall superparamagnetic iron dark cloud? J. Blood Med. 3, 77–85 (2012).

future science group www.futuremedicine.com 1015


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

70 Landry R, Jacobs PM, Davis R, Shenouda M, Bolton angiogenesis in wounds and malignant tumors. J. Leukoc.
WK. Pharmacokinetic study of ferumoxytol: a new iron Biol. 70(4), 478–490 (2001).
replacement therapy in normal subjects and hemodialysis 85 Lin EY, Li JF, Gnatovskiy L et al. Macrophages regulate the
patients. Am. J. Nephrol. 25(4), 400–410 (2005). angiogenic switch in a mouse model of breast cancer. Cancer
71 Neuwelt EA, Hamilton BE, Varallyay CG et al. Ultrasmall Res. 66(23), 11238–11246 (2006).
superparamagnetic iron oxides (USPIOs): a future alternative 86 Mantovani A, Bottazzi B, Colotta F, Sozzani S, Ruco L.
magnetic resonance (MR) contrast agent for patients at risk The origin and function of tumor-associated macrophages.
for nephrogenic systemic fibrosis (NSF)? Kidney Int. 75(5), Immunol. Today 13(7), 265–270 (1992).
465–474 (2009).
87 Bingle L, Brown NJ, Lewis CE. The role of tumour-
72 Auerbach M, Strauss W, Auerbach S, Rineer S, Bahrain associated macrophages in tumour progression: implications
H. Safety and efficacy of total dose infusion of 1,020 mg for new anticancer therapies. J. Pathol. 196(3), 254–265
of ferumoxytol administered over 15 min. Am. J. Hematol. (2002).
88(11), 944–947 (2013).
88 Nishie A, Ono M, Shono T et al. Macrophage infiltration
73 Enochs WS, Harsh G, Hochberg F, Weissleder R. Improved and heme oxygenase-1 expression correlate with angiogenesis
delineation of human brain tumors on MR images using in human gliomas. Clin. Cancer Res. 5(5), 1107–1113 (1999).
a long-circulating, superparamagnetic iron oxide agent.
89 Rossi ML, Jones NR, Candy E et al. The mononuclear cell
J. Magn. Reson. Imaging 9(2), 228–232 (1999).
infiltrate compared with survival in high-grade astrocytomas.
74 Hunt MA, Bago AG, Neuwelt EA. Single-dose contrast agent Acta Neuropathol. 78(2), 189–193 (1989).
for intraoperative MR imaging of intrinsic brain tumors by
90 Corot C, Warlin D. Superparamagnetic iron oxide
using ferumoxtran-10. Am. J. Neuroradiol. 26(5), 1084–1088
nanoparticles for MRI: contrast media pharmaceutical
(2005).
company R&D perspective. Wiley Interdiscip. Rev. Nanomed.
75 Murillo TP, Sandquist C, Jacobs PM, Nesbit G, Manninger Nanobiotechnol. 5(5), 411–422 (2013).
S, Neuwelt EA. Imaging brain tumors with ferumoxtran-10,
91 Dosa E, Guillaume DJ, Haluska M et al. Magnetic
a nanoparticle magnetic resonance contrast agent. Therapy 2,
Resonance Imaging of intracranial tumors: intra-patient
871–882 (2005).
comparison of gadoteridol and ferumoxytol. Neurooncology
76 Neuwelt EA, Varallyay P, Bago AG, Muldoon LL, Nesbit G, 13(2), 251–260 (2011).
Nixon R. Imaging of iron oxide nanoparticles by MR and
92 Manninger SP, Muldoon LL, Nesbit G, Murillo T, Jacobs
light microscopy in patients with malignant brain tumours.
PM, Neuwelt EA. An exploratory study of ferumoxtran-10
Neuropathol. Appl. Neurobiol. 30(5), 456–471 (2004).
nanoparticles as a blood-brain barrier imaging agent
77 Taschner CA, Wetzel SG, Tolnay M, Froehlich J, Merlo A, targeting phagocytic cells in CNS inflammatory lesions.
Radue EW. Characteristics of ultrasmall superparamagnetic Am. J. Neuroradiol. 26(9), 2290–2300 (2005).
iron oxides in patients with brain tumors. Am. J. Roentgenol.
93 Holness CL, Simmons DL. Molecular cloning of CD68,
185(6), 1477–1486 (2005).
a human macrophage marker related to lysosomal
78 Moore A, Marecos E, Bogdanov A Jr, Weissleder R. Tumoral glycoproteins. Blood 81(6), 1607–1613 (1993).
distribution of long-circulating dextran-coated iron oxide
94 Knauth M, Egelhof T, Roth SU, Wirtz CR, Sartor K.
nanoparticles in a rodent model. Radiology 214(2), 568–574
Monocrystalline iron oxide nanoparticles: possible
(2000).
solution to the problem of surgically induced intracranial
79 Muldoon LL, Sandor M, Pinkston KE, Neuwelt EA. contrast enhancement in intraoperative MR imaging.
Imaging, distribution, and toxicity of superparamagnetic Am. J. Neuroradiol. 22(1), 99–102 (2001).
iron oxide magnetic resonance nanoparticles in the rat brain
95 Aronen HJ, Gazit IE, Louis DN et al. Cerebral blood
and intracerebral tumor. Neurosurgery785–796 (2005).
volume maps of gliomas: comparison with tumor grade and
80 Rainov NG, Zimmer C, Chase M et al. Selective uptake of histologic findings. Radiology 191(1), 41–51 (1994).
viral and monocrystalline particles delivered intra-arterially
96 Sugahara T, Korogi Y, Kochi M et al. Correlation of MR
to experimental brain neoplasms. Hum. Gene Ther. 6(12),
imaging-determined cerebral blood volume maps with
1543–1552 (1995).
histologic and angiographic determination of vascularity of
81 Zimmer C, Wright SC Jr, Engelhardt RT et al. Tumor cell gliomas. Am. J. Roentgenol. 171(6), 1479–1486 (1998).
endocytosis imaging facilitates delineation of the glioma-
97 Gahramanov S, Muldoon LL, Varallyay CG et al.
brain interface. Exp. Neurol. 143(1), 61–69 (1997).
Pseudoprogression of glioblastoma after chemo- and
82 Kremer S, Pinel S, Vedrine PO et al. Ferumoxtran-10 radiation therapy: diagnosis by using dynamic susceptibility-
enhancement in orthotopic xenograft models of human weighted contrast-enhanced perfusion MR imaging with
brain tumors: an indirect marker of tumor proliferation? ferumoxytol versus gadoteridol and correlation with survival.
J. Neurooncol. 83(2), 111–119 (2007). Radiology 266(3), 842–852 (2013).
83 Pollard JW. Tumour-educated macrophages promote tumour • Provides important data for distinguishing treatment
progression and metastasis. Nat. Rev. Cancer 4(1), 71–78
effects from viable tumor using ferumoxytol.
(2004).
98 Barajas RF Jr, Chang JS, Segal MR et al. Differentiation
84 Crowther M, Brown NJ, Bishop ET, Lewis CE.
of recurrent glioblastoma multiforme from radiation
Microenvironmental influence on macrophage regulation of
necrosis after external beam radiation therapy with dynamic

1016 Nanomedicine (Lond.) (2015) 10(6) future science group


Iron oxides for neuro-oncology  Review

susceptibility-weighted contrast-enhanced perfusion MR 113 Batchelor TT, Sorensen AG, Di Tomaso E et al. AZD2171,
imaging. Radiology 253(2), 486–496 (2009). a pan-VEGF receptor tyrosine kinase inhibitor, normalizes
99 Thompson EM, Guillaume DJ, Dosa E et al. Dual contrast tumor vasculature and alleviates edema in glioblastoma
perfusion MRI in a single imaging session for assessment patients. Cancer Cell 11(1), 83–95 (2007).
of pediatric brain tumors. J. Neuro Oncol. 109(1), 105–114 114 Thomas AA, Omuro A. Current role of anti-angiogenic
(2012). strategies for glioblastoma. Curr. Treat. Options Oncol. 15(4),
100 Claes A, Gambarota G, Hamans B et al. Magnetic resonance 551–566 (2014).
imaging-based detection of glial brain tumors in mice after 115 Jain RK. Normalization of tumor vasculature: an emerging
antiangiogenic treatment. Int. J. Cancer 122(9), 1981–1986 concept in antiangiogenic therapy. Science 307(5706), 58–62
(2008). (2005).
101 Gahramanov S, Muldoon LL, Li X, Neuwelt EA. Improved 116 Chinot OL, Wick W, Mason W et al. Bevacizumab
perfusion MR imaging assessment of intracerebral tumor plus radiotherapy-temozolomide for newly diagnosed
blood volume and antiangiogenic therapy efficacy in a rat glioblastoma. N. Engl. J. Med. 370(8), 709–722 (2014).
model with ferumoxytol. Radiology 261(3), 796–804 (2011). 117 Gilbert MR, Dignam JJ, Armstrong TS et al. A randomized
102 Varallyay CG, Muldoon LL, Gahramanov S et al. Dynamic trial of bevacizumab for newly diagnosed glioblastoma.
MRI using iron oxide nanoparticles to assess early vascular N. Engl. J. Med. 370(8), 699–708 (2014).
effects of antiangiogenic versus corticosteroid treatment in a 118 Field KM, Jordan JT, Wen PY, Rosenthal MA, Reardon
glioma model. J. Cereb. Blood Flow Metabol. 29(4), 853–860 DA. Bevacizumab and glioblastoma: Scientific review, newly
(2009). reported updates, and ongoing controversies. Cancer doi:
103 Christoforidis GA, Yang M, Kontzialis MS et al. High 10.1002/cncr.28935 (2014) (Epub ahead of print).
resolution ultra high field Magn. Reson. Imaging of glioma 119 Emblem KE, Farrar CT, Gerstner ER et al. Vessel calibre-a
microvascularity and hypoxia using ultra-small particles of potential MRI biomarker of tumour response in clinical
iron oxide. Invest. Radiol. 44(7), 375–383 (2009). trials. Nat. Rev. Clin. Oncol. 11(10), 566–584 (2014).
104 Gambarota G, Leenders W, Maass C et al. Characterisation 120 Tropres I, Pannetier N, Grand S et al. Imaging
of tumour vasculature in mouse brain by USPIO contrast- the microvessel caliber and density: Principles and
enhanced MRI. Br. J. Cancer 98(11), 1784–1789 (2008). applications of microvascular MRI. Magn. Reson. Med.
105 Tropres I, Lamalle L, Peoc’h M et al. In vivo assessment of doi: 10.1002/mrm.25396 (2014) (Epub ahead of print).
tumoral angiogenesis. Magn. Reson. Med. 51(3), 533–541 121 Dennie J, Mandeville JB, Boxerman JL, Packard SD, Rosen
(2004). BR, Weisskoff RM. NMR imaging of changes in vascular
106 Christen T, Ni W, Qiu D et al. High-resolution cerebral morphology due to tumor angiogenesis. Magn. Reson. Med.
blood volume imaging in humans using the blood pool 40(6), 793–799 (1998).
contrast agent ferumoxytol. Magn. Reson. Med. 70(3), 122 Lemasson B, Valable S, Farion R, Krainik A, Remy C,
705–710  (2013).  Barbier EL. In vivo imaging of vessel diameter, size, and
107 Varallyay CG, Nesbit E, Fu R et al. High-resolution density: a comparative study between MRI and histology.
steady-state cerebral blood volume maps in patients with Magn. Reson. Med. 69(1), 18–26 (2013).
central nervous system neoplasms using ferumoxytol, a 123 Pannetier N, Lemasson B, Christen T et al. Vessel size index
superparamagnetic iron oxide nanoparticle. J. Cereb. Blood measurements in a rat model of glioma: comparison of the
Flow Metabol. 33(5), 780–786 (2013). dynamic (Gd) and steady-state (iron-oxide) susceptibility
108 Gahramanov S, Varallyay C, Tyson RM et al. Diagnosis of contrast MRI approaches. NMR Biomed. 25(2), 218–226
pseudoprogression using MRI perfusion in patients with (2012).
glioblastoma multiforme may predict improved survival. 124 Bouchet A, Lemasson B, Le Duc G et al. Preferential effect
CNS Oncol. 3(6), 389–400 (2014). of synchrotron microbeam radiation therapy on intracerebral
109 Weller M, Stupp R, Reifenberger G et al. MGMT promoter 9L gliosarcoma vascular networks. Int. J. Radiat. Oncol. Biol.
methylation in malignant gliomas: ready for personalized Phys. 78(5), 1503–1512 (2010).
medicine? Nat. Rev. Neurol. 6(1), 39–51 (2010). 125 Kostourou V, Robinson SP, Whitley GS, Griffiths
110 Nanegrungsunk D, Onchan W, Chattipakorn N, JR. Effects of overexpression of dimethylarginine
Chattipakorn SC. Current evidence of temozolomide and dimethylaminohydrolase on tumor angiogenesis assessed
bevacizumab in treatment of gliomas. Neurol. Res. 37(2), by susceptibility magnetic resonance imaging. Cancer Res.
167–183 (2014). 63(16), 4960–4966 (2003).
111 ClinicalTrials.gov. Assessing dynamic magnetic resonance 126 Quarles CC, Schmainda KM. Assessment of the
(MR) imaging in patients with recurrent high grade glioma morphological and functional effects of the anti-angiogenic
receiving chemotherapy. agent SU11657 on 9L gliosarcoma vasculature using dynamic
http://clinicaltrials.gov  susceptibility contrast MRI. Magn. Reson. Med. 57(4),
112 Lemasson B, Christen T, Tizon X et al. Assessment of 680–687 (2007).
multiparametric MRI in a human glioma model to monitor 127 Serduc R, Christen T, Laissue J et al. Brain tumor vessel
cytotoxic and anti-angiogenic drug effects. NMR Biomed. response to synchrotron microbeam radiation therapy: a short-
24(5), 473–482 (2011). term in vivo study. Phys. Med. Biol. 53(13), 3609–3622 (2008).

future science group www.futuremedicine.com 1017


Review  Iv, Telischak, Feng, Holdsworth, Yeom & Daldrup-Link

128 Emblem KE, Mouridsen K, Bjornerud A et al. Vessel delivery and targeted therapy of glioblastoma. Cancer Res.
architectural imaging identifies cancer patient responders to 70(15), 6303–6312 (2010).
anti-angiogenic therapy. Nat. Med. 19(9), 1178–1183 (2013). 139 Aboody KS, Najbauer J, Metz MZ et al. Neural stem cell-
129 Sorensen AG, Batchelor TT, Zhang WT et al. A “vascular mediated enzyme/prodrug therapy for glioma: preclinical
normalization index” as potential mechanistic biomarker to studies. Sci. Transl. Med. 5(184), 184ra159 (2013).
predict survival after a single dose of cediranib in recurrent 140 Gutova M, Shackleford GM, Khankaldyyan V et al. Neural
glioblastoma patients. Cancer Res. 69(13), 5296–5300 stem cell-mediated CE/CPT-11 enzyme/prodrug therapy in
(2009). transgenic mouse model of intracerebellar medulloblastoma.
130 Batchelor TT, Gerstner ER, Emblem KE et al. Improved Gene Ther. 20(2), 143–150 (2013).
tumor oxygenation and survival in glioblastoma patients 141 Gutova M, Frank JA, D’apuzzo M et al. Magn. Reson.
who show increased blood perfusion after cediranib and Imaging tracking of ferumoxytol-labeled human neural stem
chemoradiation. Proc. Natl Acad. Sci. USA 110(47), cells: studies leading to clinical use. Stem Cells Transl. Med.
19059–19064 (2013). 2(10), 766–775 (2013).
131 Kioi M, Vogel H, Schultz G, Hoffman RM, Harsh 142 Daldrup-Link HE, Rudelius M, Oostendorp RA et al.
GR, Brown JM. Inhibition of vasculogenesis, but not Targeting of hematopoietic progenitor cells with MR contrast
angiogenesis, prevents the recurrence of glioblastoma after agents. Radiology 228(3), 760–767 (2003).
irradiation in mice. J. Clin. Invest. 120(3), 694–705 (2010).
143 Wankhede M, Bouras A, Kaluzova M, Hadjipanayis CG.
132 Ahn GO, Brown JM. Matrix metalloproteinase-9 is required Magnetic nanoparticles: an emerging technology for
for tumor vasculogenesis but not for angiogenesis: role of malignant brain tumor imaging and therapy. Expert Rev.
bone marrow-derived myelomonocytic cells. Cancer Cell Clin. Pharmacol. 5(2), 173–186 (2012).
13(3), 193–205 (2008).
144 Weinstein JS, Varallyay CG, Dosa E et al. Superparamagnetic
133 Martin BJ. Inhibiting vasculogenesis after radiation: a new iron oxide nanoparticles: diagnostic Magn. Reson. Imaging
paradigm to improve local control by radiotherapy. Semin. and potential therapeutic applications in neurooncology and
Radiat. Oncol. 23(4), 281–287 (2013). central nervous system inflammatory pathologies, a review.
134 Orringer DA, Koo YE, Chen T, Kopelman R, Sagher J. Cereb. Blood Flow Metabol. 30(1), 15–35 (2010).
O, Philbert MA. Small solutions for big problems: the •• Provides excellent review of iron oxide nanoparticles in
application of nanoparticles to brain tumor diagnosis and
brain tumor imaging.
therapy. Clin. Pharmacol. Ther. 85(5), 531–534 (2009).
145 Maier-Hauff K, Ulrich F, Nestler D et al. Efficacy and safety
135 Reddy GR, Bhojani MS, Mcconville P et al. Vascular
of intratumoral thermotherapy using magnetic iron-oxide
targeted nanoparticles for imaging and treatment of brain
nanoparticles combined with external beam radiotherapy
tumors. Clin. Cancer Res. 12(22), 6677–6686 (2006).
on patients with recurrent glioblastoma multiforme.
136 Cai W, Shin DW, Chen K et al. Peptide-labeled near-infrared J. Neurooncol. 103(2), 317–324 (2011).
quantum dots for imaging tumor vasculature in living
146 Tani N, Joly O, Iwamuro H et al. Direct visualization of non-
subjects. Nano Lett. 6(4), 669–676 (2006).
human primate subcortical nuclei with contrast-enhanced
137 Madhankumar AB, Slagle-Webb B, Mintz A, Sheehan JM, high field MRI. Neuroimage 58(1), 60–68 (2011).
Connor JR. Interleukin-13 receptor-targeted nanovesicles are
147 Burrell JS, Walker-Samuel S, Baker LC et al. Evaluation
a potential therapy for glioblastoma multiforme. Mol. Cancer
of novel combined carbogen USPIO (CUSPIO) imaging
Ther. 5(12), 3162–3169 (2006).
biomarkers in assessing the antiangiogenic effects of
138 Hadjipanayis CG, Machaidze R, Kaluzova M et al. cediranib (AZD2171) in rat C6 gliomas. Int. J. Cancer
EGFRvIII antibody-conjugated iron oxide nanoparticles for 131(8), 1854–1862 (2012).
magnetic resonance imaging-guided convection-enhanced

1018 Nanomedicine (Lond.) (2015) 10(6) future science group

View publication stats

Potrebbero piacerti anche