ArticlePDF AvailableLiterature Review

Review of tracer kinetic models in evaluation of gliomas using dynamic contrast-enhanced imaging

Frontiers
Frontiers in Oncology
Authors:

Abstract and Figures

Glioma is the most common type of primary malignant tumor of the central nervous system (CNS), and is characterized by high malignancy, high recurrence rate and poor survival. Conventional imaging techniques only provide information regarding the anatomical location, morphological characteristics, and enhancement patterns. In contrast, advanced imaging techniques such as dynamic contrast-enhanced (DCE) MRI or DCE CT can reflect tissue microcirculation, including tumor vascular hyperplasia and vessel permeability. Although several studies have used DCE imaging to evaluate gliomas, the results of data analysis using conventional tracer kinetic models (TKMs) such as Tofts or extended-Tofts model (ETM) have been ambiguous. More advanced models such as Brix’s conventional two-compartment model (Brix), tissue homogeneity model (TH) and distributed parameter (DP) model have been developed, but their application in clinical trials has been limited. This review attempts to appraise issues on glioma studies using conventional TKMs, such as Tofts or ETM model, highlight advancement of DCE imaging techniques and provides insights on the clinical value of glioma management using more advanced TKMs.
This content is subject to copyright.
Review of tracer kinetic
models in evaluation of
gliomas using dynamic
contrast-enhanced imaging
Jianan Zhou
1,2,3
, Zujun Hou
4
, Chuanshuai Tian
1,2,3
,
Zhengyang Zhu
2,3
, Meiping Ye
2,3
, Sixuan Chen
2,3
,
Huiquan Yang
2,3
, Xin Zhang
1,2,3
*and Bing Zhang
1,2,3
1
Department of Radiology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical
University, Nanjing, China,
2
Institute of Medical Imaging and Articial Intelligence, Nanjing University,
Nanjing, China,
3
Medical Imaging Center, Department of Radiology, Nanjing Drum Tower Hospital,
Afliated Hospital of Medical School, Nanjing University, Nanjing, China,
4
The Jiangsu Key Laboratory
of Medical Optics, Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of
Sciences, Suzhou, China
Glioma is the most common type of primary malignant tumor of the central
nervous system (CNS), and is characterized by high malignancy, high recurrence
rate and poor survival. Conventional imaging techniques only provide
information regarding the anatomical location, morphological characteristics,
and enhancement patterns. In contrast, advanced imaging techniques such as
dynamic contrast-enhanced (DCE) MRI or DCE CT can reect tissue
microcirculation, including tumor vascular hyperplasia and vessel permeability.
Although several studies have used DCE imaging to evaluate gliomas, the results
of data analysis using conventional tracer kinetic models (TKMs) such as Tofts or
extended-Tofts model (ETM) have been ambiguous. More advanced models
such as Brixs conventional two-compartment model (Brix), tissue homogeneity
model (TH) and distributed parameter (DP) model have been developed, but their
application in clinical trials has been limited. This review attempts to appraise
issues on glioma studies using conventional TKMs, such as Tofts or ETM model,
highlight advancement of DCE imaging techniques and provides insights on the
clinical value of glioma management using more advanced TKMs.
KEYWORDS
glioma, dynamic contrast-enhanced, tracer kinetic model, diagnosis,
treatment response
Frontiers in Oncology frontiersin.org01
OPEN ACCESS
EDITED BY
Michael Albert Thomas,
University of California, Los Angeles,
United States
REVIEWED BY
Johannes Kerschbaumer,
Innsbruck Medical University, Austria
Vera Catharina Keil,
VU Medical Center, Netherlands
*CORRESPONDENCE
Xin Zhang
neuro_zx@163.com
RECEIVED 02 February 2024
ACCEPTED 29 May 2024
PUBLISHED 14 June 2024
CITATION
Zhou J, Hou Z, Tian C, Zhu Z, Ye M,
Chen S, Yang H, Zhang X and Zhang B (2024)
Review of tracer kinetic models in
evaluation of gliomas using dynamic
contrast-enhanced imaging.
Front. Oncol. 14:1380793.
doi: 10.3389/fonc.2024.1380793
COPYRIGHT
©2024Zhou,Hou,Tian,Zhu,Ye,Chen,Yang,
Zhang and Zhang. This is an open-access
article distributed under the terms of the
Creative Commons Attribution License (CC BY).
The use, distribution or reproduction in other
forums is permitted, provided the original
author(s) and the copyright owner(s) are
credited and that the original publication in
this journal is cited, in accordance with
accepted academic practice. No use,
distribution or reproduction is permitted
which does not comply with these terms.
TYPE Review
PUBLISHED 14 June 2024
DOI 10.3389/fonc.2024.1380793
1 Introduction
Glioma originates from the neurostromal cells and is the most
common primary tumor of the central nervous system (CNS) (1). It
is characterized by wide-spread invasion and angiogenesis, with
short median survival duration and high recurrence rate (2). First-
line therapy for gliomas consists of radiotherapy, surgery,
concomitant chemoradiotherapy and adjuvant chemotherapy with
temozolomide (3), while immunotherapies are currently in the pre-
clinical and clinical stages of testing (4). The treatment response of
glioma is primarily evaluated on the basis of contrast enhanced T1-
weighted magnetic resonance imaging (MRI). However, the
correlation between changes in enhancement and the treatment
response is often confounded by the presence of radiation necrosis,
pseudoprogression or pseudoresponse (57)(Figures 1 and 2for
example), thereby warranting more advanced imaging techniques
for accurate assessment.
Dynamic contrast enhancement (DCE) imaging is a non-
invasive approach that can provide in vivo physiological and
metabolic information of tissues, and assess microvascular
features such as the degree of vascularity and disruption of
vascular wall permeability (811). DCE imaging data can be
analyzed in terms of both semi-quantitative and quantitative
parameters. The former includes time-intensity curve (TIC)
parameters, such as initial area under the curve (IAUC) and time
to peak, which are easy to derive (12) but challenging to reproduce
across studies due to differences in data acquisition and subject
conditions. DCE imaging data can be quantitatively analyzed using
a tracer kinetic model (TKM), a mathematical description of tracer
molecular transport within the tissue microenvironment that
derives quantitative values of various model parameters
pertaining to the tissue status. Several clinical studies have tested
DCE imaging for various applications, including glioma assessment
(1317). Most of these studies used Tofts or extended-Tofts model
(ETM), which represents early development in DCE imaging. More
advanced TKMs have been developed (18,19), but have received
less attention in clinical studies.
In this review, the evaluation of glioma using conventional
TKMs, advances in DCE imaging techniques, and the clinical
potential of advanced TKMs in glioma management have
been discussed.
2 Materials and methods
2.1 Literature search and selection strategy
We searched for candidate articles describing the different
TKMs for gliomas in PubMed and Science Direct databases
published between January 1950 and May 2024. The search
strategy of key terms used was ((tracer kinetic model OR
pharmacokinetic model) OR (Tofts OR generalized kinetic
model) OR extended Tofts model OR (two-compartment model
OR two-compartment exchange model) OR tissue homogeneity
model OR distributed parameter model)) AND (glioma OR
glioblastoma) AND (dynamic contrast-enhanced OR DCE). The
studies were included based on the following inclusion criteria: (1)
clinical studies that employed DCE data in patients with gliomas, or
experimental animal studies that refer to pathophysiological
mechanism of microenvironment; (2) original research published
in English with the full text available; and (3) the theoretical basis of
tracer kinetic model and its application in glioma diagnosis and
evaluation after treatment. The following types of studies were
excluded: (1) unrelated or irrelevant studies, such as those that did
not employ DCE techniques to investigated gliomas; and (2) studies
focusing on other topics that are irrelevant to our research purpose.
After a detailed evaluation and screening,105 studies that met our
criteria were included and reviewed. The article selection process is
shown as a owchart in Figure 3.
2.2 Fundamental concepts and primary
tracer kinetic models
Tracer kinetic models describe the transport of tracer molecular
within the tissue microenvironment. The physical space of the
movement of tracer molecules in the tissue is termed as
compartment. Typically, there are two well-dened compartments
ABC
FIGURE 1
A 59-year-old female with glioma of the left parieto-occipital lobe, treated with surgery and radiation therapy. (A) MR image before surgery; (B) 10
months after surgery and completion of radiation therapy showed enhancing lesion; (C) follow-up MR showed resolution of the lesion.
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org02
in the eld, namely, the compartment of intravascular plasma space
(IVPS), and the compartment of extravascular extracellular space
(EES). Furthermore, based on the distribution of the tracer, a
compartment in TKMs can be categorized as homogeneous or non-
homogeneous. For a homogeneous compartment, diffusive resistances
are assumed to be zero and the tracer is assumed to move fast and
distribute instantaneously and evenly upon arrival in the
compartment, which is also termed as a well-mixed compartment.
Since the distribution of the tracer is uniform in a well-mixed or
homogeneous compartment, the tracer concentration is constant in
space and only changes with time (18). TKMs with assumptions on
homogeneous or well-mixed compartment are often named as
lumped parameter models. In contrast, the tracer concentration
varies in a non-homogeneous compartment, and is therefore a
function of both space and time. In general, a homogeneous or
well-mixed compartment would simplify the modeling process and
computation. Thus, the early TKMs were developed based on this
assumption. However, the uniform distribution of a tracer in the
compartment would depend on rapid movement of tracer molecules
or sufciently long measurement time, neither of which is viable in
clinical practice. Therefore, subsequent TKMs introduced a
concentration gradient in space to account for the variation in
tracer distribution, leading to the assumption of non-homogeneous
compartment. Nevertheless, tracer concentration in these models is
homogeneous in the radial direction and variable in the axial direction.
The meaning of these terms has been summarized in Table 1.
Primary tracer kinetic models, including Tofts, ETM, Brixs
conventional two-compartment model (Brix), tissue homogeneity
model (TH), and distributed parameter model (DP), have been
listed in Table 2. It is worth pointing out that the notation of a TKM
could be different in different studies, and what is introduced here
follows largely the notations in earlier review papers on technical
aspects of tracer kinetic modeling (18,19). Tofts model was also
named as generalized kinetic (GK) model in (18). The model
developed by Brix and coauthors (24,25) has been denoted as
two-compartment exchange model (2CXM) in some literature (19,
35,36). However, this notation reects also the fundamental
features of other models such as TH and DP, likely leading to
confusion in understanding the connection and the difference
among these TKMs. To emphasize that the exchange between
two-compartments is the common feature of these models, the
notation of the model proposed by Brix and coauthors is denoted as
ABCDEF
FIGURE 2
A 67-year-old male with WHO grade 2 glioma of the left basal ganglia and posterior horn of lateral ventricle, treated with surgery, radiotherapy plus
concomitant and adjuvant temozolomide. (A) MR image before surgery; (B) MR image after surgery; (C) 1 month after surgery; (D) 10 months after
surgery; (E) 13 months after surgery; (F) 15 months after surgery. Follow-up examinations demonstrated the presence of enhancing lesion in 10
months after surgery, which expanded in 13 months and reduced in 15 months.
FIGURE 3
Flowchart of the literature screening process.
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org03
conventional two-compartment model (CC or CC2) in (18,37), or
as Brix model in (38). Since ETM is also of two-compartment by
nature, confusion could be arisen between CC and ETM. For clarity
and simplicity, this presentation adopts the notation of Brix model.
For completeness, the equations of the models are given
as follows:
Tofts model
Ctiss(t) = AIF Ktransexp Ktrans
ve
t

where denotes the convolution operator.
Extended-Tofts model (ETM)
Ctiss(t) = AIF Ktransexp Ktrans
ve
t

+vp

Brixs conventional two-compartment (Brix) model, Equations 1a1c
Ctiss(t) = AIF Fp½Aexp(at)+(1A)exp(bt)(1a)
where
a
b
!
=1
2PS
vp
+PS
ve
+Fp
vp
!
±ffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
PS
vp
+PS
ve
+Fp
vp
!
2
4PS
ve
Fp
vp
v
u
u
t
2
43
5
(1b)
A=
a+PS
vp+PS
ve
ab (1c)
Distributed parameter (DP) model, Equation 2
Ctiss(t) = AIF
Fp
u(t)utvp
Fp

+
utvp
Fp

1exp PS
Fp

1+ tvp
Fp
0exp PS
ve
t

ffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
PS
ve
PS
Fp
1
t
sI12ffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
PS
ve
PS
Fp
t
s
!
dt
"#()
8
>
>
>
<
>
>
>
:
9
>
>
>
=
>
>
>
;
(2)
where u(t) denotes the Heaviside unit-step function and I
1
is the
modied Bessel function.
Tissue homogeneity (TH) model, Equation 3
Ctiss(t) = AIF
   Fp1exp PS
Fp
hi
exp Fp
ve1exp PS
Fp
hi
tFp
vp
nono
(3)
Tofts assumes that IVPS is signicantly smaller than EES, and
thus only involves EES. All other models are two-compartment
models. The movement of tracer molecules in tissue generally
involves intravascular transport and exchange between
intravascular and interstitial space. The former reects blood ow
(CBF) and the latter indicates permeability of endothelial wall (PS).
Tofts and ETM utilize one parameter (K
trans
) to describe both
movements, whereas Brix, TH and DP differentiate between the two
and model them separately (24). Tofts, ETM and Brix models
assume the compartment to be well-mixed. In the TH model, EES is
well-mixed but IVPS is non-homogenous. On the other hand, both
compartments are non-homogenous in the DP model. The
TABLE 1 The concept and clinical signicance of several terms.
Concept Meaning
compartment physical distribution space of tracer in the tissue
well-mixed compartment tracer distributes evenly throughout compartment
not well-
mixed compartment
tracer concentration changes with time and space
in compartment
IVPS intravascular compartment (intravascular
plasma space)
EES interstitial compartment (extravascular
extracellular space)
AIF arterial input function
VIF vascular input function
relative or
normalized parameter
parameter is normalized with respect to
contralateral healthy tissue
TABLE 2 Summary of primary tracer kinetic models.
Model References Compartment Transport
rate
parameter
Well-
mixed
assumption
Independent
parameters
Derived
parameters
Tofts [Kety et al. (20),Tofts et al. (21)] EES K
trans
well-mixed K
trans
,V
e
K
ep
ETM [Tofts et al. (22)] EES, IVPS K
trans
well-mixed K
trans
,V
e
,V
p
K
ep
Brix [Hayton et al. (23), Brix et al. (24),
Brix et al. (25), Larsson et al. (26)]
EES, IVPS CBF, PS well-mixed CBF, PS, V
e
,V
p
MTT, E
TH [Johnson (27)], Lawrence et al.
(28), Lawrence et al. (29), Lee
et al. (30)]
EES, IVPS CBF, PS well-mixed EES, not
well-mixed IVPS
CBF, PS, V
e
,V
p
MTT, E
DP [Bassingthwaighte et al. (31),
Larson et al. (32), Koh et al. (33),
Koh et al. (34)]
EES, IVPS CBF, PS not well-mixed in
both compartments
CBF, PS, V
e
,V
p
MTT, E
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org04
parameters derived from these models are listed in Table 2. The
technical details pertaining to these models have been
comprehensively discussed in previous reviews (18,19). The
kinetic parameters were graphically presented in Figure 4.
2.3 Comparison between DCE MRI and
DCE CT
As a well-established imaging technique, DCE can be
performed by data acquisition on either MRI or CT scanners,
followed by image analysis using TKMs (39). Each imaging
modality has its merits and demerits. CT is fast in scanning and
allows acquisition of images with high resolution in both spatial and
temporal space, but at cost of X-ray radiation. MRI is advantageous
in better contrast in soft tissue and radiation-free. To reduce
radiation in DCE CT, non-uniform acquisition strategy can be
adopted to decrease the number of X-ray exposure in the design of
DCE CT protocol, with frequent scans during artery phase and less
frequent scans during delayed phase. A key difference in terms of
DCE language between two modalities lies in the calculation of
contrast concentration. Contrast concentration in DCE CT follows
simply a linear relationship with CT image signal, whereas the
relationship between contrast concentration in DCE MRI with MRI
signal is much more complicated, which relates to changes in T1
values of tissue before and after contrast injection. A practical
approach to estimating T1 value of tissue is the method of variable
ip angles and the computation involves several MR scanning
factors such as time of repetition (TR), time of echo (TE), ip
angle, homogeneity of B1 eld. After deriving contrast
concentration from either CT or MR image signals, the analytical
process of concentration-time curve will be exactly the same
between DCE CT and DCE MRI. For a good appraisal on DCE
CT and DCE MRI, interested readers can refer to the review paper
(39), where it was shown that data acquisition and analysis were
well comparable despite inherent differences in signal production
and mechanism of tissue contrast enhancement.
3 Application of conventional tracer
kinetic models in glioma evaluation
3.1 Application in glioma grading
Various studies have analyzed the relationship between DCE
imaging parameters and glioma grading (4049). Santarosa et al.
(50) used ETM of DCE MRI in a cohort of 26 glioma patients and
demonstrated that V
p
and K
trans
differed signicantly between low-
grade and high-grade gliomas. Using the same model, Zhang et al.
(51) found that K
trans
and V
e
values calculated in 28 glioma patients
based on DCE MRI increased with advanced tumor grade, and
signicant differences were observed between the low (I and II) and
high (III and IV) grade gliomas, as well as between grades II and III.
Awasthi et al. (52) applied Tofts model of DCE MRI to 76 glioma
patients and showed that K
ep
and V
e
could differentiate low-grade
from high-grade tumors, although there was no signicant
correlation between K
trans
and the expression of MMP-9, which
plays a key role in the disruption of the blood-brain barrier (BBB)
by degrading extracellular matrix in order to facilitate tumor cell
inltration and metastasis. In contrast, other studies (53,54) based
on DCE MRI have shown that K
trans
is the most effective parameter
for differentiating between glioma grades. These contradictory
ndings can be attributed to the differences in the extent of BBB
disruption among different tumor grades, as well as the different
mechanisms underlying BBB disruption in infectious and
neoplastic pathologies.
3.2 Correlation with
immunohistochemical markers
Since the 2021 World Health Organization (WHO) guidelines
on the histological classication of central nervous system tumors
were published (55), molecular markers have been instrumental in
the diagnosis of gliomas. For instance, IDH mutation, 1p/19q co-
deletion, TERT promoter mutation, and EGFR gene amplication
are key biomarkers used for the classication of diffuse gliomas in
FIGURE 4
The graphic illustration of various kinetic parameters in tracer kinetic models. IVPS, intravascular compartment (intravascular plasma space); EES,
interstitial compartment (extravascular extracellular space); K
trans
, transfer constant; K
ep
, washout rate; CBF, tumor blood ow; V
e
, interstitial volume;
V
p
, blood volume; PS, permeabilitysurface area product.
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org05
adults, and DCE TKMs have been used for evaluating the status of
these markers in glioma patients (52,5663).
IDH mutation is associated with a survival benet in glioma
patients (64). Wang et al. (65) retrospectively studied the IDH
mutation status of 30 patients with low grade gliomas (LGGs) using
ETM of DCE MRI, and showed that K
trans
,V
e
and V
p
were higher
values in the IDH wild-type compared to the IDH mutant LGGs. In
contrast, Brendle et al. (66) reported that DCE MRI kinetic
parameters derived using the same model did not distinguish
between IDH mutant and wild-type astrocytomas. Ahn et al. (67)
retrospectively studied the molecular markers in 132 LGG patients
using ETM of DCE MRI, and found no signicant difference in the
DCE kinetic parameters between the IDH mutant and wild-
type gliomas.
MGMT promoter methylation predicts favorable prognosis
after alkylating drug-based chemotherapy in patients with IDH-
wild-type glioblastomas (68). Several studies have evaluated the
correlation between MGMT promoter methylation and DCE
kinetic parameters in gliomas (6971). Ahn et al. (67) showed
that K
trans
,V
e
and V
p
values derived using ETM of DCE MRI were
signicantlylowerinMGMTmethylatedLGGsthaninthe
unmethylated counterparts. Another study (70) using the Tofts
model of DCE MRI showed that K
trans
values were signicantly
higher in the MGMT methylated tumors, while K
ep
and V
e
showed
no signicant difference between gliomas with methylated and
unmethylated MGMT. Hilario et al. (69) analyzed 49 glioma
patients using ETM of DCE MRI, and did not detect any
signicant differences in the DCE kinetic parameters of the
MGMT methylated and non-methylated tumors. Zhang et al. (71)
further showed that gliomas with non-methylated MGMT had
higher V
e
and K
trans
values with ETM of DCE MRI than those
with methylated MGMT.
3.3 Differential diagnosis of glioma, PCNSL,
and metastasis
Due to differences in clinical treatment and prognosis, it is
critical to distinguish between primary central nervous system
lymphoma (PCNSL), high-grade gliomas (HGGs), and metastatic
glioma (7279). Xi et al. (80) used Tofts model of DCE MRI to
retrospectively analyze 8 cases of PCNSL, 21 cases of HGGs and 6
cases of metastasis, and detected signicantly higher K
trans
and V
e
in
the PCNSL tumors compared to HGGs and metastatic tumors.
However, Kickingereder et al. (81) did not observe any signicant
difference in the V
e
values of PCNSL and GBM using ETM of DCE
MRI. Lin et al. (82) showed that PCNSL had higher values of K
trans
using ETM of DCE MRI compared to GBM, although the difference
was not statistically signicant. Jin et al. (83) used ETM of DCE
MRI to show that K
trans
had the highest specicity and sensitivity in
differentiating between GBM, PCNSL, and metastasis. Kang et al.
(84) calculated the V
p
in glioblastomas and PCNSL through ETM of
DCE MRI, and found that the values were signicantly higher in the
former. In contrast, Abe et al. (43) used ETM of DCE MRI to
analyze 29 lesions (including glioma, metastatic tumor and
lymphoma) and found that V
p
was not helpful in differentiating
PCNSL from GBM.
3.4 Evaluation of treatment response
All patients with glioblastoma eventually relapse. It is
challenging to differentiate relapsed tumor from other treatment-
related changes during the follow-up of glioma patients (8592).
Thomas et al. (93) found that V
p
and K
trans
measurements were
lower in glioblastoma patients with pseudoprogression compared to
those with relapsed lesions using ETM of DCE MRI. Yun et al. (94)
also applied ETM of DCE MRI to differentiate false progression
from true progression, and found that K
trans
and V
e
were
signicantly higher in the latter, whereas V
p
value was similar for
both types of lesions. Jing et al. (95) applied the Tofts model
of DCE MRI to a retrospective analysis of 51 patients with
new enhancement lesions after standard radiotherapy and
chemotherapy after surgical resection, and found no signicant
difference in V
e
and K
ep
between the pseudoprogression group and
the recurrence group.
4 Assessment of gliomas using
advanced tracer kinetic models
4.1 Glioma grading
Jain et al. (96,97) investigated glioma grading using perfusion
CT (PCT) with the TH model (27) to estimate permeability and
blood volume. While the rCBV increased more than the PS in
LGGs, grade 3 and especially grade 4 gliomas showed a greater
increase in PS compared to rCBV. The rate of change in the rCBV/
PS ratio may correlate to changes occurring at the tumor
microvasculature level. Both PS and CBV were higher in the
HGGs compared to the LGGs. Furthermore, PS can also be used
to differentiate WHO grade 3 glioma from grade 4 glioma. Tietze
et al. (35) presented a Bayesian framework for parameter
optimization of tracer kinetic modeling and compared the Brix
model and ETM in the grading of 42 untreated glioma patients.
Brix-derived V
p
showed the best diagnostic performance with AUC
of 0.97, followed by ETM-derived K
trans
with an AUC of 0.92.
However, the diagnostic performance of permeability parameter
was low, with AUC of 0.57.
4.2 Monitoring of treatment response
4.2.1 In vivo human studies
Jensen et al. (98) correlated parameters derived from a gamma
capillary transit time model of DCE MRI, which is based on the
distributed capillary approximated TH model (99,100), with
molecular markers of hypoxia, vascularity, proliferation, and
progression-free and overall survival (OS) in a cohort of 18
glioma patients. The derived parameters included tumor blood
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org06
ow (CBF), extraction fraction (E), permeabilitysurface area
product (PS), transfer constant (K
trans
), washout rate (K
ep
),
interstitial volume (V
e
), blood volume (V
p
), capillary transit time
(tc), and capillary heterogeneity (a1). The study showed that a1,
tc, K
ep
, and V
p
were correlated with HIF-1aand VEGF expression,
whereas V
e
and a1were correlated with OS. The other imaging
markers were not helpful in predicting OS. In particular, none of the
blood ow and permeability parameters (K
trans
, CBF, E, PS) showed
any correlation with patient outcome.
Lundemann et al. (101) explored the feasibility of predicting
tumor recurrence using multi-modal imaging based on DCE MRI at
the pre-radiotherapy stage in a cohort of 16 glioblastoma patients
using Brix-derived DCE parameters. The median MTT, V
p
,V
e
and
PS derived from scans prior to chemoradiotherapy showed
differences between recurring and non-recurring voxels.
Henriksen et al. (102) investigated the diagnostic value of Brix-
derived parameters of DCE MRI for short-term disease progression
in 60 anaplastic astrocytoma and glioblastoma patients with
suspected recurrence and evaluable outcome within 6 months of
follow-up as determined by histopathology, MRI ndings or clinical
decision. The blood volume and vascular permeability were
signicantly higher in the progressive lesions compared to the
non-progressive lesions. ROC analysis showed that blood ow
and blood volume had AUC values of 0.76 and 0.78 respectively,
which were higher than that of vascular permeability (0.68).
Larsen et al. (103) utilized Brix of DCE MRI to differentiate tumor
recurrence from radiation necrosis in 19 glioma patients following
surgery and radiation therapy, and demonstrated that an empirical
threshold of 2 ml/100 g for blood volume allowed detection of
regressing lesions with sensitivity and specicity of 100% each. In
comparison, neither blood ow nor permeability parameter could
discriminate between regressing and progressing lesions.
4.2.2 Ex vivo animal studies
Kiser et al. (36) applied the Brix model of DCE MRI to evaluate
test-retest repeatability and tumor response of a murine
glioblastoma model at 7 T to a combination therapy of
bevacizumab and uorouracil. Test-retest experiments
demonstrated that there was no signicant difference between the
scans in terms of the median values of parameters, except for K
trans
.
The compartmental volume fractions (V
e
and V
p
) remained more
consistent between scans while the vascular functional parameters
(CBF and PS) showed noticeable increase in values, likely due to
physiological changes in the tumor between scans. The tumor
volume in the control and treated groups did not differ
signicantly at any time point, which corresponded to similar
tracer kinetic parameters in both groups.
Yeung et al. (104) investigated the efcacy of CT perfusion
imaging as an early biomarker of the response to stereotactic
radiosurgery in a rat glioma model using the TH model (27). Rats
with orthotopic C6 glioma tumors received either mock irradiation
or stereotactic radiosurgery delivered by Helical Tomotherapy. The
responders to stereotactic radiosurgery showed lower relative CBV,
and PS on day 7 post-stereotactic radiosurgery when compared to
controls and non-responders. Relative CBV and PS on day 7 were
correlated with the OS, and predicted survival with 92% accuracy.
4.3 Correlation to
immunohistochemical markers
Li et al. (105) applied the DP model of DCE MRI to a dataset
consisting of 55 glioma patients to assess glioma isocitrate
dehydrogenase (IDH) mutation. The IDH-mutant gliomas
showed signicantly lower CBF, PS, V
p
, E and V
e
compared to
the IDH-wildtype gliomas. V
p
exhibited the best performance in
differentiating between IDH-mutant and IDH-wildtype gliomas
(AUC=0.92), followed by PS (AUC=0.82) and E (AUC=0.8).
Furthermore, the Tofts parameters K
trans
and V
e
were lower for
the IDH-mutant gliomas, and no signicant difference was
observed in K
ep
. The AUCs of K
trans
,V
e
, and K
ep
were 0.69, 0.79,
and 0.55 respectively. These ndings suggested that IDH-mutant
gliomas have lower vascularity, vessel permeability and blood ow
compared to IDH-wildtype gliomas, which may explain the better
outcomes in patients with IDH-mutated versus IDH-
wildtype gliomas.
5 Discussion
5.1 Advantage of advanced TKMs over
conventional ones
The fth edition of the WHO Classication of CNS Tumors
(WHO CNS5), published in 2021, builds on the fourth edition
updated in 2016 and the work of the Consortium to Inform
Molecular and Practical Approaches to CNS Tumor Taxonomy,
further advancing the role of molecular diagnostics in the
classication of CNS tumors, but remains rooted in other
established methods for assessing tumor characteristics, including
histology and immunohistochemistry (55). Besides, the existing
evaluation of glioma features is still based on pathological biopsy as
the gold standard. Due to the high heterogeneity of gliomas and the
inuence of non-uniform sampling error of tumors, the diagnostic
accuracy is limited by the subjective judgment of pathologists. In
2021 WHO CNS5, EGFR amplication is commonly seen in IDH
wild-type gliomas. EGFR is a tyrosine kinase receptor regulating cell
proliferation and differentiation by interacting with epidermal
growth factor (EGF) and tumor growth factor-a(TGF-a), and
can reect microvascular proliferation in tumors (106). With the
development of MRI technologies, more abundant characteristics of
tumor microenvironment have been provided for the non-invasive
diagnosis of gliomas. DCE MRI is sensitive to the destruction of the
blood-brain barrier (BBB) and is closely related to microvascular
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org07
proliferation and vascular wall permeability, which can effectively
evaluate tumor angiogenesis and obtain various kinetic parameters
reecting tissue microcirculation function (9).
The focus of this review was to reveal the characteristics of the
immune microenvironment of gliomas based on the conventional
and advanced TKMs of DCE imaging, and to provide a non-
invasive method for the diagnosis and treatment response
evaluation of gliomas. Most studies on glioma assessment with
DCE imaging have used Tofts or ETM for image postprocessing and
data analysis, and the results show considerable variability. For
instance, some studies (50,107) have reported that ETM-derived V
p
is a useful imaging parameter that can discriminate between LGG
and HGG, whereas one study (51) showed that V
e
and not V
p
differed signicantly between LGG and HGG. Likewise, Arevalo-
Perez et al. (42) showed that V
p
had the best discriminatory power
in glioma grading, whereas Jung et al. (44) reported that K
trans
was
the most signicant pharmacokinetic parameter. ETM-derived
parameters can also distinguish IDH-mutant gliomas from IDH
wildtype gliomas (65), although some studies (66,67) have reported
contradictory ndings. While ETM_V
p
was reported to be
signicantly higher for glioblastomas compared to PCNSL in one
study (84), another study (43) showed that it could not differentiate
between the two lesions. Furthermore, K
trans
derived from the Tofts
model was demonstrated to be a promising discriminatory
biomarker for LGG relative to HGG (41,108), although Awasthi
et al. (52) found that K
trans
was not signicantly different between
LGG and HGG. The variations in the results across the different
studies using Tofts or ETM, and the inconsistent performance of
K
trans
in particular, have been reported in earlier reviews (13,14).
Quantitative imaging biomarkers alliance (QIBA) has
recommended ETM for analyzing brain tumors through DCE
imaging (109). Both imaging hardware and DCE tracer kinetic
modeling have undergone signicant advances over the years,
thereby allowing acquisition of DCE images with higher temporal
resolution, better signal-to-noise ratio, wider brain coverage and
increased spatial resolution, and enabling separate quantication of
CBF and PS. The advantages of a DP model over the Tofts model
have been demonstrated in the assessment of the IDH mutation
status in glioma (105). Recent test-retest experiments using Brix
(36) showed that only the median K
trans
was signicantly different
between scans, which might justify the separate modeling of
intravascular transport and exchange between intra- and extra-
vascular space in Brix, instead of mixed modeling using one
transport rate parameter (K
trans
). HIF-1aexpression is
signicantly associated with HGGs (110) and is an excellent
biomarker for glioma grading (111). Furthermore, gliomas are
typically characterized by a marked increase in the formation of
blood vessels with abnormal blood ow and increased vessel
permeability. In fact, blood volume is a promising imaging
biomarker for glioma grading (42,50,107). As expected, V
p
derived using a DP model showed good correlation with HIF-1a
expression (r= 0.747, P=0.043) (108). However, low correlation was
observed between HIF-1aexpression and ETM-derived V
p
(r= 0.149, P=0.219) in another study (111).
Physiological interpretation of K
trans
has been ambiguous in
clinical trials using Tofts or ETM, and is generally described as a
volume transfer constant that reects vessel wall permeability (13,
14). Several theoretical descriptions have been provided to interpret
K
trans
as a combination of blood ow and vessel wall permeability
(112,113). Separate measurement of CBF and PS has been well
addressed in nuclear medicine (114), which is crucial to the
understanding of tissue hemodynamics, and has spurned the
development of more advanced DCE TKMs. In Figure 5,the
parametric maps of ETM and DP for a glioblastoma (GBM)
patient after surgery have been compared. The follow-up MR
images in Row 1 demonstrated the evolution of enhancing lesion.
DP-derived parameters in Row 2 showed that the enhancing lesion
was characterized by reduced blood ow, increase in PS and
marginal increase in V
e
compared to the contralateral tissue.
ETM-derived parameters in Row 2 manifested as reduced K
trans
,
marginal increase in V
p
, and increase in V
e
. As previously reported
(24), V
e
in the brain tumor reects the transfer of tracer molecules
between intravascular and interstitial space due to insufcient
leakage from the intravascular space and attainment of steady
levels in the interstitial space during the insufcient scanning
period. Hence, ETM and DP illustrated similar pattern in vessel
wall permeability in this case. Nonetheless, the lower value of K
trans
,
which is generally explained as reduced vessel wall permeability,
may be indicative of reduced blood ow for this case.
The MR images of WHO grade 4 glioma at the right frontal lobe
treated with surgery and chemoradiotherapy are shown in Figure 6.
Small enhancing lesions appeared in the 31
th
month of follow-up
after surgery and continued to grow. DCE scan was performed in
the 37
th
month of follow-up, and analyzed using the distributed
parameter model. The parametric maps are shown in the second
row, which indicate reduced blood ow and increased vessel
permeability in the lesion compared to the contralateral tissue.
The patient underwent a second surgery, and histopathological
analysis revealed necrotic foci with no evidence of recurrent tumor.
In the traditional WHO classication of tumors, each tumor
type has a unique WHO grading, such as anaplastic astrocytoma,
which is automatically assigned to WHO grade III. The latest 2021
WHO Classication of CNS Tumors emphasizes the combination
of histological grades and molecular markers, such as Glioblastoma,
IDH-wild type; Astrocytoma, IDH-mutant; or Oligodendroglioma,
IDH-mutant & 1p/19q codeleted. IDH mutant astrocytoma
expands from the CNS in WHO grade 24. However, most of the
current studies are based on the 2016 WHO Classication, lacking
specic tumor pathological classication diagnosis (Table 3).
Therefore, future studies need to use more advanced MRI
technologies for further comprehensive diagnosis and stratied
reporting of gliomas.
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org08
5.2 Standardization of DCE data acquisition
and postprocessing
Despite limited research on advanced DCE imaging TKMs for
glioma, the differences across studies are obvious. Brix-derived
blood volume has shown high discriminatory ability for
regressing lesions, whereas blood ow and permeability
parameters showed fairly low discriminating power in the
differential diagnosis of tumor recurrence (103). In contrast, a
recent study (102) demonstrated similar ability of blood ow and
blood volume in differentiating progressive lesions from non-
progressive lesions, and signicantly higher permeability in the
former. Multiple factors can contribute to these discrepancies,
such as DCE imaging protocols, data post-processing, scanners,
operators in data processing, sample size and patient characteristics,
etc. QIBA has made the following recommendations to standardize
DCE MR imaging protocol (115): (1) 3D T1-weighted GRE
sequence, (2) variable ip angle (VFA) method to estimate T1
map and contrast agent concentration, (3) same sequence for pre-
contrast VFA scan and post-contrast dynamic scan, (4) temporal
resolution not lower than 4 s in most cases, (5) sufciently long
scanning duration of about 6 min for permeability measurement.
The DCE protocols used in earlier studies often deviated from
QIBA recommendations, whereas the more recent studies (102)
have protocols closer to QIBA guidelines. DCE imaging data is
largely analyzed using commercial software programs that differ in
algorithm implementation, which may result in signicant
differences in the estimated DCE parameters (116). When
analyzing DCE images, it is critical to accurately determine the
contrast concentration from image intensities. The relationship
FIGURE 5
59-year-old male with glioblastoma of the left temporal lobe. Row 1: follow-up MR examinations after surgery showed the evolution of enhancing
lesion. Row 2: parametric maps of blood ow CBF, vessel permeability PS and fractional volume of interstitial space V
e
as derived using DP.
Parametric maps of volume transfer constant K
trans
,V
p
and V
e
as derived using ETM.
ABCDEF
FIGURE 6
35-year-old female with WHO grade 4 glioma of the right frontal lobe. Postoperative pathology conrmed the enhanced lesion as radiation necrosis
(RN). Row 1: follow-up MR examinations after surgery showed the presence and evolution of enhancing lesion. (A) MR image after the rst surgery;
(B) 30 months after surgery; (C)31 months after surgery; (D)33 months after surgery; (E)35 months after surgery; (F)37 months after surgery. Row 2:
parametric maps of blood ow CBF, mean transit time MTT, fractional volume of intravascular space CBV, fractional volume of interstitial space V
e
,
vessel permeability PS, and extraction ratio E as derived using DP model.
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org09
between image intensity and concentration can be non-linear,
particular for MRI. In some software programs, linear assumption
between change in signal intensity and gadolinium concentration is
used to directly derive the contrast concentration-time curve from
signal intensity-time curve (42). The choice of vascular input
function (VIF) also affects the estimated values of DCE
parameters. It would be ideal to select VIF from DCE images for
each patient. Some software programs use population VIF or
empirical VIF, as summarized in the QIBA prole (109).
Standardization of imaging protocol and postprocessing
procedure is crucial in making DCE reproducible and bringing
forward the technology into clinical practice.
5.3 Cross-validation in DCE studies
ROC curve analysis is a useful tool for characterizing the
differential diagnostic ability of potential biomarkers, which can be
quantied by calculating the area under the ROC curve (AUC), or by
nding an optimal threshold from the ROC curve and determining the
associated sensitivity, specicity and accuracy. Most studies on DCE
imaging-based glioma assessment directly applied the ROC curve
method to the total data, and reported the performance of DCE
parameters. In clinical practice, however, the imaging data is
invariably subjected to different sources of perturbations. Therefore,
the resulting diagnostic metrics could be biased towards best matching
pattern in the current data, and their performance could be
signicantly different when applied to new data. This phenomenon is
called overtting in machine learning (117) and is particularly acute
whenthesamplesizeissmall,whichisoftenthecaseingliomastudies.
This issue is commonly resolved by cross-validation, which is typically
performed by dividing data into the training set and validation set, and
applying the information derived from the former to the latter. K-fold
cross-validation is the most frequently used cross-validation method in
machine learning. However, the leave-one-out cross-validation method
is recommended for glioma studies since the cohort size is usually
small. Shao et al. (38) used this method to assess the parameters of DCE
imaging in a small cohort of cervical cancer patients. The following
steps were used: (1) one data was left out, and the remaining data was
used as the training set to build a model and make prediction on the
excluded data, (2) the previous step was repeated for each data, and (3)
the differential diagnostic ability was quantied by summarizing all
predicted values. In the current DCE glioma studies, performance
quantication without cross-validation has resulted in variation
between the results of different research groups.
6 Conclusion
DCE imaging is effective in glioma grading and therapeutic
effect monitoring, and its parameters are potential imaging markers
for glioma diagnosis. However, the discrepancies between the
ndings in different studies warrant further improvement and
validation of this technique with standardization of protocol
design, and data post-processing in multi-center and large
cohorts. Advanced DCE imaging techniques that allow separate
modeling of blood ow and vessel permeability have advantages
over conventional counterparts, although more studies are needed
to ascertain the clinical value.
Author contributions
JZ: Conceptualization, Formal analysis, Funding acquisition,
Investigation, Writing original draft, Writing review & editing.
ZH: Conceptualization, Formal analysis, Methodology, Software,
Writing original draft, Writing review & editing. CT:
Conceptualization, Formal analysis, Writing original draft,
Writing review & editing. ZZ: Conceptualization, Formal
analysis, Writing original draft, Writing review & editing.
TABLE 3 Application of tracer kinetic models in glioma grading and molecular markers according to WHO classication.
Model References Tasks WHO Classication
Tofts
Zhang et al. (2012) (51), Awasthi et al. (2012) (52) grading 2007
Ahn et al. (2014) (70) MGMT methylation 2007
Jia et al. (2021) (41), Zhao et al. (2017) (53) grading 2016
ETM
Santarosa et al. (2016) (50) grading 2016
Wang et al. (2020) (65), Brendle et al. (2018) (66) IDH mutation 2016
Ahn et al. (2023) (67) IDH mutation, EGFR, MGMT and TERT 2016
Hilario et al. (2019) (69) IDH mutation, ATRX, and MGMT 2016
Zhang et al. (2020) (71) IDH mutation, MGMT and TERT 2016
Anzalone et al. (2018) (40) grading, IDH mutation and 1p19q codeletion 2016
TH Jain et al. (2008,2015) (96,97) grading 2007
DP Li et al. (2020) (105) IDH mutation 2016
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org10
MY: Conceptualization, Formal analysis, Writing original draft,
Writing review & editing. SC: Conceptualization, Writing
original draft, Writing review & editing. HY: Conceptualization,
Writing original draft, Writing review & editing. XZ:
Conceptualization, Funding acquisition, Methodology,
Supervision, Writing original draft, Writing review & editing.
BZ: Funding acquisition, Supervision, Writing original draft,
Writing review & editing.
Funding
The author(s) declare nancial support was received for the
research, authorship, and/or publication of this article. This
research was funded by the National Science and Technology
Innovation 2030 Major program of Brain Science and Brain-
Like Research(2022ZD0211800); the National Natural Science
Foundation of China (82271965, 82302172); General Project
Supported by Medical Science and technology development
Foundation, Nanjing Department of Health (YKK22083,
YKK23103), the Jiangsu Funding Program for Excellent
Postdoctoral Talent (2022ZB694); and fundings for Clinical Trials
from the Afliated Drum Tower Hospital, Medical School of
Nanjing University (2022-LCYJ-PY-15, 2022-LCYJ-MS-03, 2021-
LCYJ-PY-36, 2022-LCYJ-MS-25, 2021-LCYJ-PY-20); Postgraduate
Research & Practice Innovation Program of Jiangsu
Province (JX22014155).
Conict of interest
The authors declare that the research was conducted in the
absence of any commercial or nancial relationships that could be
construed as a potential conict of interest.
Publishers note
All claims expressed in this article are solely those of the authors
and do not necessarily represent those of their afliated
organizations, or those of the publisher, the editors and the
reviewers. Any product that may be evaluated in this article, or
claim that may be made by its manufacturer, is not guaranteed or
endorsed by the publisher.
References
1. Ostrom QT, Gittleman H, Truitt G, Boscia A, Kruchko C, Barnholtz-Sloan JS.
CBTRUS statistical report: primary brain and other central nervous system tumors
diagnosed in the United States in 20112015. Neuro Oncol. (2018) 20:iv1iv86.
doi: 10.1093/neuonc/noy131
2. Lapointe S, Perry A, Butowski NA. Primary brain tumours in adults. Lancet.
(2018) 392:43246. doi: 10.1016/S01406736(18)309905
3. Weller M, van den Bent M, Preusser M, Le Rhun E, Tonn JC, Minniti G, et al.
EANO guidelines on the diagnosis and treatment of diffuse gliomas of adulthood. Nat
Rev Clin Oncol. (2021) 18:17086. doi: 10.1038/s41571020-00447-z
4. Lim M, Xia Y, Bettegowda C, Weller M. Current state of immunotherapy for
glioblastoma. Nat Rev Clin Oncol. (2018) 15:42242. doi: 10.1038/s41571018-00035
5. Dhermain FG, Hau P, Lanfermann H, Jacobs AH, van den Bent MJ. Advanced
MRI and PET imaging for assessment of treatment response in patients with gliomas.
Lancet Neurol. (2010) 9:90620. doi: 10.1016/S14744422(10)701812
6. Zikou A, Sioka C, Alexiou GA, Fotopoulos A, Voulgaris S, Argyropoulou MI.
Radiation necrosis, pseudoprogression, pseudoresponse, and tumor recurrence:
imaging challenges for the evaluation of treated gliomas. Contrast Media Mol
Imaging. (2018) 2018:6828396. doi: 10.1155/2018/6828396
7. Fink J, Born D, Chamberlain MC. Pseudoprogression: relevance with respect to
treatment of high-grade gliom as. Curr Treat Optio ns Oncol. (2011) 12:24052.
doi: 10.1007/s11864011-01571
8. Heye AK, Thrippleton MJ, Armitage PA, ValdesHerna
ndez MDC, Makin SD, Glatz A,
et al. Tracer kinetic modelling for DCE-MRI quantication of subtle blood-brain barrier
permeability. Neuroimage. (2016) 125:44655. doi: 10.1016/j.neuroimage.2015.10.018
9. Heye AK, Culling RD, Valdes Hernandez MDC, Thrippleton MJ, Wardlaw JM.
Assessment of blood-brain barrier disruption using dynamic contrast-enhanced MRI.
A systematic review. NeuroImage Clin. (2014) 6:26274. doi: 10.1016/j.nicl.2014.09.002
10. Nielsen T, Wittenborn T, Horsman MR. Dynamic contrast-enhanced magnetic
resonance imaging (DCE-MRI) in preclinical studies of antivascular treatments.
Pharmaceutics. (2012) 4:56389. doi: 10.3390/pharmaceutics4040563
11. Eilaghi A, Yeung T, dEsterre C, Bauman G, Yartsev S, Easaw J, et al. Quantitative
perfusion and permeability biomarkers in brain cancer from tomographic CT and MR
images. biomark Cancer. (2016) 8:4759. doi: 10.4137/BIC.S31801
12. Narang J, Jain R, Arbab AS, Mikkelsen T, Scarpace L, Rosenblum ML, et al.
Differentiating treatment-induced necrosis from recurrent/progressive brain tumor using
nonmodel-based semiquantitative indices derived from dynamic contrast-enhanced T1-
weighted MR perfusion. Neuro Oncol. (2011) 13:103746. doi: 10.1093/neuonc/nor075
13. Zahra MA, Hollingsworth KG, Sala E, Lomas DJ, Tan LT. Dynamic contrast-
enhanced MRI as a predictor of tumour response to radiotherapy. Lancet Oncol. (2007)
8:6374. doi: 10.1016/S14702045(06)710129
14. OConnor JPB, Jackson A, Parker GJM, Roberts C, Jayson GC. Dynamic
contrast-enhanced MRI in clinical trials of antivascular therapies. Nat Rev Clin
Oncol. (2012) 9:16777. doi: 10.1038/nrclinonc.2012.2
15. Fouke SJ, Benzinger T, Gibson D, Ryken TC, Kalkanis SN, Olson JJ. The role of
imaging in the management of adults with diffuse low grade glioma: A systematic
review and evidence-based clinical practice guideline. J Neurooncol. (2015) 125:45779.
doi: 10.1007/s11060015-19089
16. Guida L, Stumpo V, Bellomo J, van Niftrik CHB, Sebök M, Berhouma M, et al.
Hemodynamic imaging in cerebral diffuse glioma-partA:concept,differential
diagnosis and tumor grading. Cancers (Basel). (2022) 14:1432. doi: 10.3390/
cancers14061432
17. Hirschler L, Sollmann N, Schmitz-Abecassis B, Pinto J, Arzanforoosh F, Barkhof
F, et al. Advanced MR techniques for preoperative glioma characterization: part 1.
J Magn Reson Imaging. (2023) 57:165575. doi: 10.1002/jmri.28662
18. Koh TS, Bisdas S, Koh DM, Thng CH. Fundamentals of tracer kinetics for
dynamic contrast-enhanced MRI. JMagnResonImaging. (2011) 34:126276.
doi: 10.1002/jmri.22795
19. Sourbron SP, Buckley DL. Tracer kinetic modelling in MRI: estimating perfusion
and capillary permeability. Phys Med Biol. (2012) 57:R133. doi: 10.1088/0031-9155/
57/2/R1
20. Kety SS. The theory and applications of the exchange of inert gas at the lungs and
tissues. Pharmacol Rev. (1951) 3:141.
21. Tofts PS, Kermode AG. Measurement of the blood-brain barrier permeability
and leakage space using dynamic MR imaging. 1. Fundamental concepts. Magn Reson
Med. (1991) 17:35767. doi: 10.1002/mrm.1910170208
22. Tofts PS. Modeling tracer kinetics in dynamic Gd-DTPA MR imaging. J Magn
Reson Imaging. (1997) 7:91101. doi: 10.1002/jmri.1880070113
23. Hayton P, Brady M, Tarassenko L, Moore N. Analysis of dynamic MR breast
images using a model of contrast enhancement. Med Image Anal. (1997) 1:20724.
doi: 10.1016/s13618415(97)850116
24. Brix G, Bahner ML, Hoffmann U, Horvath A, Schreiber W. Regional blood
ow, capillary permeability, and compartmental volumes: measurement with
dynamic CTinitial experience. Radiology. (1999) 210:26976. doi: 10.1148/
radiology.210.1.r99ja46269
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org11
25. Brix G, Kiessling F, Lucht R, Darai S, Wasser K, Delorme S, et al.
Microcirculation and microvasculature in breast tumors: pharmacokinetic analysis of
dynamic MR image series. Magn Reson Med. (2004) 52:4209. doi: 10.1002/mrm.20161
26. Larsson HBW, Courivaud F, Rostrup E, Hansen AE. Measurement of brain
perfusion, blood volume, and blood-brain barrier permeability, using dynamic
contrast-enhanced T(1)-weighted MRI at 3 tesla. Magn Reson Med. (2009) 62:1270
81. doi: 10.1002/mrm.22136
27. Johnson JA, Wilson TA. A model for capillary exchange. Am J Physiol. (1966)
210:1299303. doi: 10.1152/ajplegacy.1966.210.6.1299
28. St Lawrence KS, Lee TY. An adiabatic approximation to the tissue homogeneity
model for water exchange in the brain: I. Theoretical derivation. J Cereb Blood Flow
Metab. (1998) 18:136577. doi: 10.1097/0000464719981200000011
29. St Lawrence KS, Lee TY. An adiabatic approximation to the tissue homogeneity
model for water exchange in the brain: II. Experimental validation. J Cereb Blood Flow
Metab. (1998) 18:137885. doi: 10.1097/0000464719981200000012
30. Lee T-Y, Purdie TG, Stewart E. CT imaging of angiogenesis. Q J Nucl Med.
(2003) 47:17187.
31. Bassingthwaighte JB. A concurrent ow model for extraction during
transcapillary passage. Circ Res. (1974) 35:483503. doi: 10.1161/01.res.35.3.483
32. Larson KB, Markham J, Raichle ME. Tracer-kinetic models for measuring
cerebral blood ow using externally detected radiotracers. J Cereb Blood Flow Metab.
(1987) 7:44363. doi: 10.1038/jcbfm.1987.88
33. Koh TS, Cheong LH, Hou Z, Soh YC. A physiologic model of capillary-tissue
exchange for dynamic contrast-enhanced imaging of tumor microcirculation. IEEE
Trans BioMed Eng. (2003) 50:15967. doi: 10.1109/TBME.2002.807657
34. Koh TS, Thng CH, Lee PS, Hartono S, Rumpel H, Goh BC, et al. Hepatic
metastases: in vivo assessment of perfusion parameters at dynamic contrast-enhanced
MR imaging with dual-input two-compartment tracer kinetics model. Radiology.
(2008) 249:30720. doi: 10.1148/radiol.2483071958
35. Tietze A, Nielsen A, Klærke Mikkelsen I, Bo Hansen M, Obel A, Østergaard L,
et al. Bayesian modeling of Dynamic Contrast Enhanced MRI data in cerebral glioma
patients improves the diagnostic quality of hemodynamic parameter maps. PloS One.
(2018) 13:e0202906. doi: 10.1371/journal.pone.0202906
36. Kiser K, Zhang J, Das AB, Tranos JA, Wadghiri YZ, Kim SG. Evaluation of
cellular water exchange in a mouse glioma model using dynamic contrast-enhanced
MRI with two ip angles. Sci Rep. (2023) 13:3007. doi: 10.1038/s41598023-299911
37. Lu Y, Peng W, Song J, Chen T, Wang X, Hou Z, et al. On the potential use of
dynamic contrast-enhanced (DCE) MRI parameters as radiomic features of cervical
cancer. Med Phys. (2019) 46:5098109. doi: 10.1002/mp.13821
38. Shao J, Zhang Z, Liu H, Song Y, Yan Z, Wang X, et al. DCE-MRI
pharmacokinetic parameter maps for cervical carcinoma prediction. Comput Biol
Med. (2020) 118:103634. doi: 10.1016/j.compbiomed.2020.103634
39. OConnor JPB, Tofts PS, Miles KA, Parkes LM, Thompson G, Jackson A.
Dynamic contrast-enhanced imaging techniques: CT and MRI. Br J Radiol. (2011) 84
Spec No 2:S112120. doi: 10.1259/bjr/55166688
40. Anzalone N, Castellano A, Cadioli M, Conte GM, Cuccarini V, Bizzi A, et al.
Brain gliomas: multicenter standardized assessment of dynamic contrast-enhanced and
dynamic susceptibility contrast MR images. Radiology. (2018) 287:93343.
doi: 10.1148/radiol.2017170362
41. Jia L, Wu X, Wan Q, Wan L, Jia W, Zhang N. Effects of artery input function on
dynamic contrast-enhanced MRI for determining grades of gliomas. Br J Radiol. (2021)
94:20200699. doi: 10.1259/bjr.20200699
42. Arevalo-Perez J, Peck KK, Young RJ, Holodny AI, Karimi S, Lyo JK. Dynamic
contrast-enhanced perfusion MRI and diffusion-weighted imaging in grading of
gliomas. J Neuroimaging. (2015) 25:7928. doi: 10.1111/jon.12239
43. Abe T, Mizobuchi Y, Nakajima K, Otomi Y, Irahara S, Obama Y, et al. Diagnosis
of brain tumors using dynamic contrast-enhanced perfusion imaging with a short
acquisition time. Springerplus. (2015) 4:88. doi: 10.1186/s40064015-08616
44. Jung SC, Yeom JA, Kim J-H, Ryoo I, Kim SC, Shin H, et al. Glioma: Application
of histogram analysis of pharmacokinetic parameters from T1-weighted dynamic
contrast-enhanced MR imaging to tumor grading. AJNR Am J Neuroradiol. (2014)
35:110310. doi: 10.3174/ajnr.A3825
45. van Santwijk L, Kouwenberg V, Meijer F, Smits M, Henssen D. A systematic
review and meta-analysis on the differentiation of glioma grade and mutational status
by use of perfusion-based magnetic resonance imaging. Insights Imaging. (2022)
13:102. doi: 10.1186/s13244022-012307
46. Okuchi S, Rojas-Garcia A, Ulyte A, Lopez I, UsinskieneJ, Lewis M, et al.
Diagnostic accuracy of dynamic contrast-enhanced perfusion MRI in stratifying
gliomas: A systematic review and meta-analysis. Cancer Med. (2019) 8:556473.
doi: 10.1002/cam4.2369
47. Abrigo JM, Fountain DM, Provenzale JM, Law EK, Kwong JS, Hart MG, et al.
Magnetic resonance perfusion for differentiating low-grade from high-grade gliomas at
rst presentation. Cochrane Database Syst Rev. (2018) 1:CD011551. doi: 10.1002/
14651858.CD011551.pub2
48. Liang J, Liu D, Gao P, Zhang D, Chen H, Shi C, et al. Diagnostic values of DCE-
MRI and DSC-MRI for differentiation between high-grade and low-grade gliomas: A
comprehensive meta-analysis. Acad Radiol. (2018) 25:33848. doi: 10.1016/
j.acra.2017.10.001
49. Zhang J, Liu H, Tong H, Wang S, Yang Y, Liu G, et al. Clinical applications of
contrast-enhanced perfusion MRI techniques in gliomas: recent advances and current
challenges. Contrast Media Mol Imaging. (2017) 2017:7064120. doi: 10.1155/2017/
7064120
50. Santarosa C, Castellano A, Conte GM, Cadioli M, Iadanza A, Terreni MR, et al.
Dynamic contrast-enhanced and dynamic susceptibility contrast perfusion MR
imaging for glioma grading: Preliminary comparison of vessel compartment and
permeability parameters using hotspot and histogram analysis. Eur J Radiol. (2016)
85:114756. doi: 10.1016/j.ejrad.2016.03.020
51. Zhang N, Zhang L, Qiu B, Meng L, Wang X, Hou BL. Correlation of volume
transfer coefcient Ktrans with histopathologic grades of gliomas. J Magn Reson
Imaging. (2012) 36:35563. doi: 10.1002/jmri.23675
52. Awasthi R, Rathore RKS, Soni P, Sahoo P, Awasthi A, Husain N, et al.
Discriminant analysis to classify glioma grading using dynamic contrast-enhanced
MRI and immunohistochemical markers. Neuroradiology. (2012) 54:20513.
doi: 10.1007/s00234-011-0874-y
53. Zhao M, Guo L-L, Huang N, Wu Q, Zhou L, Zhao H, et al. Quantitative analysis
of permeability for glioma grading using dynamic contrast-enhanced magnetic
resonance imaging. Oncol Lett. (2017) 14:541826. doi: 10.3892/ol.2017.6895
54. Sun S, Qian H, Li F, Li Z, Wu X. [Diagnostic value of combining permeability
with T1 perfusion parameters in quantitative dynamic contrast-enhanced magnetic
resonance imaging for glioma grading]. Zhongguo Yi Xue Ke Xue Yuan Xue Bao. (2015)
37:67480. doi: 10.3881/j.issn.1000503X.2015.06.007
55. Louis DN, Perry A, Wesseling P, Brat DJ, Cree IA, Figarella-Branger D, et al. The
2021 WHO classication of tumors of the central nervous system: a summary. Neuro
Oncol. (2021) 23:123151. doi: 10.1093/neuonc/noab106
56. Hu Y, Chen Y, Wang J, Kang JJ, Shen DD, Jia ZZ. Non-invasive estimation of
glioma IDH1 mutation and VEGF expression by histogram analysis of dynamic
contrast-enhanced MRI. Front Oncol. (2020) 10:593102. doi: 10.3389/fonc.2020.593102
57. Keil VC, Gielen GH, Pintea B, Baumgarten P, Datsi A, Hittatiya K, et al. DCE-
MRI in glioma, inltration zone and healthy brain to assess angiogenesis: A biopsy
study. Clin Neuroradiol. (2021) 31:104958. doi: 10.1007/s00062021-010153
58. Ozturk K, Soylu E, Tolunay S, Narter S, Hakyemez B. Dynamic contrast-
enhanced T1-weighted perfusion magnetic resonance imaging identies glioblastoma
immunohistochemical biomarkers via tumoral and peritumoral approach: A pilot
study. World Neurosurg. (2019) 128:e195208. doi: 10.1016/j.wneu.2019.04.089
59. Li S-H, Shen N-X, Wu D, Zhang J, Zhang J-X, Jiang J-J, et al. A comparative
study between tumor blood vessels and dynamic contrast-enhanced MRI for
identifying isocitrate dehydrogenase gene 1 (IDH1) mutation status in glioma. Curr
Med Sci. (2022) 42:6507. doi: 10.1007/s11596022-2563-y
60. Park YW, Ahn SS, Park CJ, Han K, Kim EH, Kang S-G, et al. Diffusion and
perfusion MRI may predict EGFR amplication and the TERT promoter mutation
status of IDH-wildtype lower-grade gliomas. Eur Radiol. (2020) 30:647584.
doi: 10.1007/s00330020-070903
61. Hu Y, Zhang N, Yu MH, Zhou XJ, Ge M, Shen DD, et al. Volume-based
histogram analysis of dynamic contrast-enhanced MRI for estimation of gliomas IDH1
mutation status. Eur J Radiol. (2020) 131:109247. doi: 10.1016/j.ejrad.2020.109247
62. Di N, Cheng W, Jiang X, Liu X, Zhou J, Xie Q, et al. Can dynamic contrast-
enhanced MRI evaluate VEGF expression in brain glioma? An MRI-guided stereotactic
biopsy study. J Neuroradiol. (2019) 46:18692. doi: 10.1016/j.neurad.2018.04.008
63. Di N, Yao C, Cheng W, Ren Y, Qu J, Wang B, et al. Correlation of dynamic
contrast-enhanced MRI derived volume transfer constant with histological angiogenic
markers in high-grade gliomas. J Med Imaging Radiat Oncol. (2018). doi: 10.1111/
17549485.12701
64. Beiko J, Suki D, Hess KR, Fox BD, Cheung V, Cabral M, et al. IDH1 mutant
Malignant astrocytomas are more amenable to surgical resection and have a survival
benet associated with maximal surgical resection. Neuro Oncol. (2014) 16:8191.
doi: 10.1093/neuonc/not159
65. Wang X, Cao M, Chen H, Ge J, Suo S, Zhou Y. Simplied perfusion fraction from
diffusion-weighted imaging in preoperative prediction of IDH1 mutation in WHO
grade II-III gliomas: comparison with dynamic contrast-enhanced and intravoxel
incoherent motion MRI. Radiol Oncol. (2020) 54:30110. doi: 10.2478/raon-20200037
66. Brendle C, Hempel J-M, Schittenhelm J, Skardelly M, Tabatabai G, Bender B,
et al. Glioma grading and determination of IDH mutation status and ATRX loss by
DCE and ASL perfusion. Clin Neuroradiol. (2018) 28:4218. doi: 10.1007/s00062017-
0590-z
67. Ahn SH, Ahn SS, Park YW, Park CJ, Lee S-K. Association of dynamic
susceptibility contrast- and dynamic contrast-enhanced magnetic resonance imaging
parameters with molecular marker status in lower-grade gliomas: A retrospective study.
Neuroradiol J. (2023) 36:4958. doi: 10.1177/19714009221098369
68. Chamberlain MC. Prognostic or predictive value of MGMT promoter
methylation in gliomas depends on IDH1 mutation. Neurology. (2014) 82:21478.
doi: 10.1212/01.wnl.0000451452.30826.6b
69. Hilario A, Hernandez-Lain A, Sepulveda JM, Lagares A, Perez-Nuñez A, Ramos
A. Perfusion MRI grading diffuse gliomas: Impact of permeability parameters on
molecular biomarkers and survival. Neurocirugia (Astur : Engl Ed). (2019) 30:118.
doi: 10.1016/j.neucir.2018.06.004
70. Ahn SS, Shin N-Y, Chang JH, Kim SH, Kim EH, Kim DW, et al. Prediction of
methylguanine methyltransferase promoter methylation in glioblastoma using
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org12
dynamic contra st-enhanced magnetic reso nance and diffusion te nsor imaging. J
Neurosurg. (2014) 121:36773. doi: 10.3171/2014.5.JNS132279
71. Zhang H-W, Lyu G-W, He W-J, Lei Y, Lin F, Wang M-Z, et al. DSC and DCE
histogram analyses of glioma biomarkers, including IDH, MGMT, and TERT, on
differentiation and survival. Acad Radiol. (2020) 27:e26371. doi: 10.1016/
j.acra.2019.12.010
72. Suh CH, Kim HS, Jung SC, Park JE, Choi CG, Kim SJ. MRI as a diagnostic
biomarker for differentiating primary central nervous system lymphoma from
glioblastoma: A systematic review and meta-analysis. J Magn Reson Imaging. (2019)
50:56072. doi: 10.1002/jmri.26602
73. Chen X, Xie T, Fang J, Xue W, Tong H, Kang H, et al. Quantitative in vivo
imaging of tissue factor expression in glioma using dynamic contrast-enhanced MRI
derived parameters. Eur J Radiol. (2017) 93:23642. doi: 10.1016/j.ejrad.2017.06.006
74. Jung BC, Arevalo-Perez J, Lyo JK, Holodny AI, Karimi S, Young RJ, et al.
Comparison of glioblastomas and brain metastases using dynamic contrast-enhanced
perfusion MRI. J Neuroimaging. (2016) 26:2406. doi: 10.1111/jon.12281
75. Zhang H-W, Lyu G-W, He W-J, Lei Y, Lin F, Feng Y-N, et al. Differential
diagnosis of central lymphoma and high-grade glioma: dynamic contrast-enhanced
histogram. Acta Radiol. (2020) 61:12217. doi: 10.1177/0284185119896519
76. Parvaze PS, Bhattacharjee R, Verma YK, Singh RK, Yadav V, Singh A, et al.
Quantication of Radiomics features of Peritumoral Vasogenic Edema extracted from
uid-attenuated inversion recovery images in glioblastoma and isolated brain
metastasis, using T1-dynamic contrast-enhanced Perfusion analysis. NMR BioMed.
(2023) 36:e4884. doi: 10.1002/nbm.4884
77. Wang B, Wang Z, Jia Y, Zhao P, Han G, Meng C, et al. Water exchange detected
by shutter speed dynamic contrast enhanced-MRI help distinguish solitary brain
metastasis from glioblastoma. Eur J Radiol. (2022) 156:110526. doi: 10.1016/
j.ejrad.2022.110526
78. Lu S, Wang S, Gao Q, Zhou M, Li Y, Cao P, et al. Quantitative evaluation of
diffusion and dynamic contrast-enhanced magnetic resonance imaging for
differentiation between primary central nervous system lymphoma and glioblastoma.
J Comput Assist Tomogr. (2017) 41:898903. doi: 10.1097/RCT.0000000000000622
79. Choi YS, Lee H-J, Ahn SS, Chang JH, Kang S-G, Kim EH, et al. Primary central
nervous system lymphoma and atypical glioblastoma: differentiation using the initial
area under the curve derived from dynamic contrast-enhanced MR and the apparent
diffusion coefcient. Eur Radiol. (2017) 27:134451. doi: 10.1007/s00330016-44842
80. Xi Y-B, Kang X-W, Wang N, Liu T-T, Zhu Y-Q, Cheng G, et al. Differentiation
of primary central nervous system lymphoma from high-grade glioma and brain
metastasis using arterial spin labeling and dynamic contrast-enhanced magnetic
resonance imaging. Eur J Radiol. (2019) 112:5964. doi: 10.1016/j.ejrad.2019.01.008
81. Kickingereder P, Sahm F, WiestlerB, Roethke M, Heiland S, Schlemmer H-P, et al.
Evaluation of microvascular permeability with dynamic contrast-enhanced MRI for the
differentiation of primary CNS lymphoma and glioblastoma: radiologic-pathologic
correlation. AJNR Am J Neuroradiol. (2014) 35:15038. doi: 10.3174/ajnr.A3915
82. Lin X, Lee M, Buck O, Woo KM, Zhang Z, Hatzoglou V, et al. Diagnostic
accuracy of T1-weighted dynamic contrast-enhanced-MRI and DWI-ADC for
differentiation of glioblastoma and primary CNS lymphoma. AJNR Am J
Neuroradiol. (2017) 38:48591. doi: 10.3174/ajnr.A5023
83. Jin Y, Peng H, Peng J. Brain glioma localization diagnosis based on magnetic
resonance imaging. World Neurosurg. (2021) 149:32532. doi: 10.1016/
j.wneu.2020.09.113
84. Kang KM, Choi SH, Chul-Kee P, Kim TM, Park S-H, Lee JH, et al.
Differentiation between glioblastoma and primary CNS lymphoma: application of
DCE-MRI parameters based on arterial input function obtained from DSC-MRI. Eur
Radiol. (2021) 31:9098109. doi: 10.1007/s00330021-08044-z
85. Qiu J, Tao Z-C, Deng K-X, Wang P, Chen C-Y, Xiao F, et al. Diagnostic accuracy
of dynamic contrast-enhanced magnetic resonance imaging for distinguishing
pseudoprogression from glioma recurrence: a meta-analysis. Chin Med J(Engl).
(2021) 134:253543. doi: 10.1097/CM9.0000000000001445
86. Khan MN, Sharma AM, Pitz M, Loewen SK, Quon H, Poulin A, et al. High-grade
glioma management and response assessment-recent advances and current challenges.
Curr Oncol. (2016) 23:e383391. doi: 10.3747/co.23.3082
87. Artzi M, Liberman G, Nadav G, Blumenthal DT, Bokstein F, Aizenstein O, et al.
Differentiation between treatment-related changes and progressive disease in patients
with high grade brain tumors using support vector machine classication based on
DCE MRI. J Neurooncol. (2016) 127:51524. doi: 10.1007/s11060016-20557
88. Bisdas S, Naegele T, Ritz R, Dimostheni A, Pfannenberg C, Reimold M, et al.
Distinguishing recurrent high-grade gliomas from radiation injury: a pilot study using
dynamic contrast-enhanced MR imaging. Acad Radiol. (2011) 18:57583. doi: 10.1016/
j.acra.2011.01.018
89. Zakhari N, Taccone MS, Torres CH, Chakraborty S, Sinclair J, Woulfe J, et al.
Prospective comparative diagnostic accuracy evaluation of dynamic contrast-enhanced
(DCE) vs. dynamic susceptibility contrast (DSC) MR perfusion in differentiating tumor
recurrence from radiation necrosis in treated high-grade gliomas. J Magn Reson
Imaging. (2019) 50:57382. doi: 10.1002/jmri.26621
90. Fahlström M, Blomquist E, Nyholm T, Larsson E-M. Perfusion Magnetic
Resonance Imaging Changes in Normal Appearing Brain Tissue after Radiotherapy
in Glioblastoma Patients may Confound Longitudinal Evaluation of Treatment
Response. Radiol Oncol. (2018) 52:14351. doi: 10.2478/raon-20180022
91. Wang C, Sun W, Kirkpatrick J, Chang Z, Yin F-F. Assessment of concurrent
stereotactic radiosurgery and bevacizumab treatment of recurrent Malignant gliomas
using multi-modality MRI imaging and radiomics analysis. J Radiosurg SBRT. (2018)
5:17181.
92. Bressler I, Ben Bashat D, Buchsweiler Y, Aizenstein O, Limon D, Bokestein F,
et al. Model-free dynamic contrast-enhanced MRI analysis: differentiation between
active tumor and necrotic tissue in patients with glioblastoma. MAGMA. (2023) 36:33
42. doi: 10.1007/s10334022-01045-z
93. Thomas AA, Arevalo-Perez J, Kaley T, Lyo J, Peck KK, Shi W, et al. Dynamic
contrast enhanced T1 MRI perfusion differentiates pseudoprogression from recurrent
glioblastoma. J Neurooncol. (2015) 125:18390. doi: 10.1007/s11060015-1893-z
94. Yun TJ, Park C-K, Kim TM, Lee S-H, Kim J-H, Sohn C-H, et al. Glioblastoma
treated with concurrent radiation therapy and temozolomide chemotherapy:
differentiation of true progression from pseudoprogression with quantitative
dynamic contrast-enhanced MR imaging. Radiology. (2015) 274:83040.
doi: 10.1148/radiol.14132632
95. Jing H, Yan X, Li J, Qin D, Zhang N, Zhang H. The value of dynamic contrast-
enhanced magnetic resonance imaging (DCE-MRI)inthedifferentiationof
pseudoprogression and recurrence of intracranial gliomas. Contrast Media Mol
Imaging. (2022) 2022:5680522. doi: 10.1155/2022/5680522
96. Jain R, Ellika SK, Scarpace L, Schultz LR, Rock JP, Gutierrez J, et al. Quantitative
estimation of permeability surface-area product in astroglial brain tumors using
perfusion CT and correlation with histopathologic grade. AJNR Am J Neuroradiol.
(2008) 29:694700. doi: 10.3174/ajnr.A0899
97. Jain R, Grifth B, Alotaibi F, Zagzag D, Fine H, Golnos J, et al. Glioma
angiogenesis and perfusion imaging: understanding the relationship between tumor
blood volume and leakiness with increasing glioma grade. AJNR Am J Neuroradiol.
(2015) 36:20305. doi: 10.3174/ajnr.A4405
98. Jensen RL, Mumert ML, Gillespie DL, Kinney AY, Schabel MC, Salzman KL.
Preoperative dynamic contrast-enhanced MRI correlates with molecular markers of
hypoxia and vascularity in specic areas of intratumoral microenvironment and is
predictive of patient outcome. Neuro Oncol. (2014) 16:28091. doi: 10.1093/neuonc/
not148
99. Koh TS, Zeman V, Darko J, Lee TY, Milosevic MF, Haider M, et al. The inclusion
of capillary distribution in the adiabatic tissue homogeneity model of blood ow. Phys
Med Biol. (2001) 46:151938. doi: 10.1088/00319155/46/5/313
100. Schabel MC. A unied impulse response model for DCE-MRI. Magn Reson
Med. (2012) 68:163246. doi: 10.1002/mrm.24162
101. Lundemann M, Munck Af Rosenschöld P, Muhic A, Larsen VA, Poulsen HS,
Engelholm S-A, et al. Feasibility of multi-parametric PET and MRI for prediction of
tumour recurrence in patients with glioblastoma. Eur J Nucl Med Mol Imaging. (2019)
46:60313. doi: 10.1007/s00259018-41803
102. Henriksen OM, Hansen AE, Muhic A, Marner L, Madsen K, Møller S, et al.
Diagnostic yield of simultaneous dynamic contrast-enhanced magnetic resonance
perfusion measurements and [18F]FET PET in patients with suspected recurrent
anaplastic astrocytoma an d glioblastoma. Eur J Nucl Me d Mol Imaging. (2022)
49:467791. doi: 10.1007/s00259022-059173
103. Larsen VA, Simonsen HJ, Law I, Larsson HBW, Hansen AE. Evaluation of
dynamic contrast-enhanced T1-weighted perfusion MRI in the differentiation of tumor
recurrence from radiation necrosis. Neuroradiology. (2013) 55:3619. doi: 10.1007/
s00234012-11274
104. Yeung TPC, Kurdi M, Wang Y, Al-Khazraji B, Morrison L, Hoffman L, et al. CT
perfusion imaging as an early biomarker of differential response to stereotactic
radiosurgery in C6 rat gliomas. PloS One. (2014) 9:e109781. doi: 10.1371/
journal.pone.0109781
105. Li Z, Zhao W, He B, Koh TS, Li Y, Zeng Y, et al. Application of distributed
parameter model to assessment of glioma IDH mutation status by dynamic contrast-
enhanced magnetic resonance imaging. Contrast Media Mol Imaging.(2020)
2020:8843084. doi: 10.1155/2020/8843084
106. van den Bent MJ, Gao Y, Kerkhof M, Kros JM, Gorlia T, van Zwieten K, et al.
Changes in the EGFR amplication and EGFRvIII expression between paired primary
and recurrent glioblastomas. Neuro Oncol. (2015) 17:93541. doi: 10.1093/neuonc/
nov013
107. Nguyen TB, Cron GO, Mercier JF, Foottit C, Torres CH, Chakraborty S, et al.
Diagnostic accuracy of dynamic contrast-enhanced MR imaging using a phase-derived
vascular input function in the preoperative grading of gliomas. AJNR Am J Neuroradiol.
(2012) 33:153945. doi: 10.3174/ajnr.A3012
108. Choi HS, Kim AH, Ahn SS, Shin N, Kim J, Lee S-K. Glioma grading capability:
comparisons among parameters from dynamic contrast-enhanced MRI and ADC value
on DWI. Korean J Radiol. (2013) 14:48792. doi: 10.3348/kjr.2013.14.3.487
109. Shukla-Dave A, Obuchowski NA, Chenevert TL, Jambawalikar S, Schwartz LH,
Malyarenko D, et al. Quantitative imaging biomarkers alliance (QIBA)
recommendations for improved precision of DWI and DCE-MRI derived
biomarkers in multicenter oncology trials. J Magn Reson Imaging. (2019) 49:e101
21. doi: 10.1002/jmri.26518
110. Zagzag D, Zhong H, Scalzitti JM, Laughner E, Simons JW, Semenza GL.
Expression of hypoxia-inducible factor 1alpha in brain tumors: association with
angiogenesis, invasion, and progression. Cancer. (2000) 88:260618. doi: 10.1002/
(ISSN)1097-0142
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org13
111. Xie Q, Wu J, Du Z, Di N, Yan R, Pang H, et al. DCE-MRI in human gliomas: A
surrogate for assessment of invasive hypoxia marker HIF-1aBased on MRI-
neuronavigation stereotactic biopsies. Acad Radiol. (2019) 26:17987. doi: 10.1016/
j.acra.2018.04.015
112. Sourb ron SP , Buckley DL. On th e scope and inter pret ation of th e Toft s
models for DCE-MRI. Magn Reson Med. (2011) 66:73545. doi: 10.1002/
mrm.22861
113. Koh TS, Hennedige TP, Thng CH, Hartono S, Ng QS. Understanding K trans: a
simulation study based on a multiple-pathway model. Phys Med Biol. (2017) 62:N297
319. doi: 10.1088/13616560/aa70c9
114. Jacquez JA. Compartmental Analysis in Biology and Medicine.2nd ed. Ann
Arbor, MI: University of Michigan Press (1985).
115. Anon. QIBA MR Biomarker Committee. MR DCE Quantication, Quantitative
Imaging Biomarkers Alliance. Prole Stage: Public Comment. 202010-12 . Available
online at: http://qibawiki.rsna.org/index.php/Proles.
116. Goh V, Shastry M, EngledowA, Reston J, Wellsted DM, Peck J, et al. Commercial
software upgrades may signicantly alter Perfusion CT parameter values in colorectal
cancer. Eur Radiol. (2011) 21:7449. doi: 10.1007/s00330010-19674
117. Andrews JL. Addressing overtting and undertting in Gaussian model-based
clustering. Comput Stats Data Anal. (2018) 127:16071. doi: 10.1016/j.csda.2018.05.015
Zhou et al. 10.3389/fonc.2024.1380793
Frontiers in Oncology frontiersin.org14
ResearchGate has not been able to resolve any citations for this publication.
Article
Full-text available
Preoperative clinical magnetic resonance imaging (MRI) protocols for gliomas, brain tumors with dismal outcomes due to their infiltrative properties, still rely on conventional structural MRI, which does not deliver information on tumor genotype and is limited in the delineation of diffuse gliomas. The GliMR COST action wants to raise awareness about the state of the art of advanced MRI techniques in gliomas and their possible clinical translation or lack thereof. This review describes current methods, limits, and applications of advanced MRI for the preoperative assessment of glioma, summarizing the level of clinical validation of different techniques. In this first part, we discuss dynamic susceptibility contrast and dynamic contrast‐enhanced MRI, arterial spin labeling, diffusion‐weighted MRI, vessel imaging, and magnetic resonance fingerprinting. The second part of this review addresses magnetic resonance spectroscopy, chemical exchange saturation transfer, susceptibility‐weighted imaging, MRI‐PET, MR elastography, and MR‐based radiomics applications. Evidence Level: 3 Technical Efficacy: Stage 2
Article
Full-text available
Objective: The objective of this study was to determine the value of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) in assessing postoperative changes in intracranial gliomas. Method: A total of fifty-one patients who had new enhanced lesions after surgical resection followed by standard radiotherapy and chemotherapy were collected retrospectively from October 2014 to June 2021. The patients were divided into a pseudoprogression group (15 cases) and a recurrence group (36 cases) according to the pathological results of the second operation or a follow-up of more than six months. The follow-up data of all patients were complete, and DCE-MRI was performed. The images were processed to obtain the quantitative parameters K trans, Ve, and Kep and the semiquantitative parameter iAUC, which were analysed with relevant statistical software. Results: First, the difference in K trans and iAUC values between the two groups was statistically significant (P < 0.05), and the difference in Ve and Kep values was not statistically significant (P > 0.05). Second, by comparing the area under the curve, threshold, sensitivity and specificity of K trans, and iAUC, it was found that the iAUC threshold value was slightly higher than that of K trans, and the specificity of K trans was equal to that of iAUC, while the area under the curve and sensitivity of K trans were higher than those of iAUC. Third, K trans and iAUC had high accuracy in diagnosing recurrence and pseudoprogression, and K trans had higher accuracy than iAUC. Conclusion: In this study, DCE-MRI has a certain diagnostic value in the early differentiation of recurrence and pseudoprogression, offering a new method for the diagnosis and assessment of gliomas after surgery.
Article
Full-text available
Simple Summary Diffuse gliomas, and glioblastomas, in particular, represent a diagnostic and clinical challenge. Standard neuroimaging continues to have many limitations for accurate diagnostic assessment, resection planning and treatment follow-up. The present two-review series comprehensively summarizes recent evidence on hemodynamic imaging applications in the context of diffuse cerebral glioma. Part A provides an overview of the concepts underlying hemodynamic imaging modalities and critically discusses the diffuse glioma differential diagnosis and tumor grading results reported in the literature. Abstract Diffuse gliomas are the most common primary malignant intracranial neoplasms. Aside from the challenges pertaining to their treatment—glioblastomas, in particular, have a dismal prognosis and are currently incurable—their pre-operative assessment using standard neuroimaging has several drawbacks, including broad differentials diagnosis, imprecise characterization of tumor subtype and definition of its infiltration in the surrounding brain parenchyma for accurate resection planning. As the pathophysiological alterations of tumor tissue are tightly linked to an aberrant vascularization, advanced hemodynamic imaging, in addition to other innovative approaches, has attracted considerable interest as a means to improve diffuse glioma characterization. In the present part A of our two-review series, the fundamental concepts, techniques and parameters of hemodynamic imaging are discussed in conjunction with their potential role in the differential diagnosis and grading of diffuse gliomas. In particular, recent evidence on dynamic susceptibility contrast, dynamic contrast-enhanced and arterial spin labeling magnetic resonance imaging are reviewed together with perfusion-computed tomography. While these techniques have provided encouraging results in terms of their sensitivity and specificity, the limitations deriving from a lack of standardized acquisition and processing have prevented their widespread clinical adoption, with current efforts aimed at overcoming the existing barriers.
Article
Full-text available
Background:. It is crucial to differentiate accurately glioma recurrence and pseudoprogression which have entirely different prognosis and require different treatment strategies. This study aimed to assess the diagnostic accuracy of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) as a tool for distinguishing glioma recurrence and pseudoprogression. Methods:. According to particular criteria of inclusion and exclusion, related studies up to May 1, 2019, were thoroughly searched from several databases including PubMed, Embase, Cochrane Library, and Chinese biomedical databases. The quality assessment of diagnostic accuracy studies was applied to evaluate the quality of the included studies. By using the “mada” package in R, the heterogeneity, overall sensitivity, specificity, and diagnostic odds ratio were calculated. Moreover, funnel plots were used to visualize and estimate the publication bias in this study. The area under the summary receiver operating characteristic (SROC) curve was computed to display the diagnostic efficiency of DCE-MRI. Results:. In the present meta-analysis, a total of 11 studies covering 616 patients were included. The results showed that the pooled sensitivity, specificity, and diagnostic odds ratio were 0.792 (95% confidence interval [CI] 0.707–0.857), 0.779 (95% CI 0.715–0.832), and 16.219 (97.5% CI 9.123–28.833), respectively. The value of the area under the SROC curve was 0.846. In addition, the SROC curve showed high sensitivities (>0.6) and low false positive rates (
Article
Full-text available
The fifth edition of the WHO Classification of Tumors of the Central Nervous System (CNS), published in 2021, is the sixth version of the international standard for the classification of brain and spinal cord tumors. Building on the 2016 updated fourth edition and the work of the Consortium to Inform Molecular and Practical Approaches to CNS Tumor Taxonomy, the 2021 fifth edition introduces major changes that advance the role of molecular diagnostics in CNS tumor classification. At the same time, it remains wedded to other established approaches to tumor diagnosis such as histology and immunohistochemistry. In doing so, the fifth edition establishes some different approaches to both CNS tumor nomenclature and grading and it emphasizes the importance of integrated diagnoses and layered reports. New tumor types and subtypes are introduced, some based on novel diagnostic technologies such as DNA methylome profiling. The present review summarizes the major general changes in the 2021 fifth edition classification and the specific changes in each taxonomic category. It is hoped that this summary provides an overview to facilitate more in-depth exploration of the entire fifth edition of the WHO Classification of Tumors of the Central Nervous System.
Article
Full-text available
Objectives To investigate whether glioma isocitrate dehydrogenase (IDH) 1 mutation and vascular endothelial growth factor (VEGF) expression can be estimated by histogram analysis of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Methods Chinese Glioma Genome Atlas (CGGA) database was wined for differential expression of VEGF in gliomas with different IDH genotypes. The VEGF expression and IDH1 genotypes of 56 glioma samples in our hospital were assessed by immunohistochemistry. Preoperative DCE-MRI data of glioma samples were reviewed. Regions of interest (ROIs) covering tumor parenchyma were delineated. Histogram parameters of volume transfer constant (Ktrans ) and volume of extravascular extracellular space per unit volume of tissue (Ve ) derived from DCE-MRI were obtained. Histogram parameters of Ktrans , Ve and VEGF expression of IDH1 mutant type (IDH1mut ) gliomas were compared with the IDH1 wildtype (IDH1wt ) gliomas. Receiver operating characteristic (ROC) curve analysis was performed to differentiate IDH1mut from IDH1wt gliomas. The correlation coefficients were determined between histogram parameters of Ktrans , Ve and VEGF expression in gliomas. Results In CGGA database, VEGF expression in IDHmut gliomas was lower as compared to wildtype counterpart. The immunohistochemistry of glioma samples in our hospital also confirmed the results. Comparisons demonstrated statistically significant differences in histogram parameters of Ktrans and Ve [mean, standard deviation (SD), 50th, 75th, 90th. and 95th percentile] between IDH1mut and IDH1wt gliomas (P < 0.05, respectively). ROC curve analysis revealed that 50th percentile of Ktrans (0.019 min⁻¹) and Ve (0.039) provided the perfect combination of sensitivity and specificity in differentiating gliomas with IDH1mut from IDH1wt . Irrespective of IDH1 mutation, histogram parameters of Ktrans and Ve were correlated with VEGF expression in gliomas (P < 0.05, respectively). Conclusions VEGF expression is significantly lower in IDH1mut gliomas as compared to the wildtype counterpart, and it is non-invasively predictable with histogram analysis of DCE-MRI.
Article
Full-text available
Previous studies using contrast-enhanced imaging for glioma isocitrate dehydrogenase (IDH) mutation assessment showed promising yet inconsistent results, and this study attempts to explore this problem by using an advanced tracer kinetic model, the distributed parameter model (DP). Fifty-five patients with glioma examined using dynamic contrast-enhanced imaging sequence at a 3.0 T scanner were retrospectively reviewed. The imaging data were processed using DP, yielding the following parameters: blood flow F, permeability-surface area product PS, fractional volume of interstitial space Ve, fractional volume of intravascular space Vp, and extraction ratio E. The results were compared with the Tofts model. The Wilcoxon test and boxplot were utilized for assessment of differences of model parameters between IDH-mutant and IDH-wildtype gliomas. Spearman correlation r was employed to investigate the relationship between DP and Tofts parameters. Diagnostic performance was evaluated using receiver operating characteristic (ROC) curve analysis and quantified using the area under the ROC curve (AUC). Results showed that IDH-mutant gliomas were significantly lower in F ( = 0.018), PS (), Vp (), E (), and Ve ( = 0.002) than IDH-wildtype gliomas. In differentiating IDH-mutant and IDH-wildtype gliomas, Vp had the best performance (AUC = 0.92), and the AUCs of PS and E were 0.82 and 0.80, respectively. In comparison, Tofts parameters were lower in Ktrans ( = 0.013) and Ve () for IDH-mutant gliomas. No significant difference was observed in Kep ( = 0.525). The AUCs of Ktrans, Ve, and Kep were 0.69, 0.79, and 0.55, respectively. Tofts-derived Ve showed a strong correlation with DP-derived Ve (r > 0.9, ). Ktrans showed a weak correlation with F (r < 0.3, > 0.16) and a very weak correlation with PS (r < 0.06, > 0.8), both of which were not statistically significant. The findings by DP revealed a tissue environment with lower vascularity, lower vessel permeability, and lower blood flow in IDH-mutant than in IDH-wildtype gliomas, being hostile to cellular differentiation of oncogenic effects in IDH-mutated gliomas, which might help to explain the better outcomes in IDH-mutated glioma patients than in glioma patients of IDH-wildtype. The advantage of DP over Tofts in glioma DCE data analysis was demonstrated in terms of clearer elucidation of tissue microenvironment and better performance in IDH mutation assessment. 1. Introduction As the most common primary tumor in the brain, diffuse glioma arises from the glial cells which provide support functions to neurons and presents with high morbidity and variable outcomes [1]. The 2016 World Health Organization (WHO) classification of tumors of the central nervous system included well-established molecular signatures, such as isocitrate dehydrogenase (IDH) mutation status, expression of the transcription regulator ATRX, and 1p/19q codeletion status [2], where IDH is a small molecule protein involved in a number of cellular processes, including mitochondrial oxidative phosphorylation, glutamine metabolism, lipogenesis, glucose sensing, and regulation of cellular redox status [3–5]. IDH gene mutation testing is an important prognostic biomarker in gliomas and is relevant for glioma patient management and glioma stratification [6, 7]. Previous studies showed that gene expression can significantly affect the disease course, and gliomas of IDH-wildtype appear to rapidly acquire multiple complex genetic alterations and become glioblastomas very early in their development, and glioma patients with mutant IDH had significantly longer overall survival than patients without IDH mutation [6–10]. The prognostic importance of IDH mutation is independent of other known prognostic factors, including age, grade, and MGMT methylation status [6]. Hence, IDH mutations could serve as an ideal target of therapy, and imaging parameters are highly potential to capture the biologic complexity underlying molecular phenotypes in gliomas. However, conventional methods for assessment of IDH mutation status are through stereotactic biopsy invasive and prone to sampling error [11, 12]. Recently, noninvasive detection of IDH mutation status using functional imaging methods has received increasing attention [13–25]. In addition to tissue anatomic structure, functional imaging measures tissue microenvironment and provides in vivo physiologic information about brain tumors. An important functional imaging method is contrast-enhanced magnetic resonance imaging (MRI), which includes T1-weighted dynamic contrast-enhanced imaging (DCE) and T2-weighted dynamic susceptibility contrast-enhanced imaging (DSC), both of which have been applied to IDH mutation assessment in gliomas [20–25]. The tissue microenvironment of frequent study includes tumor vascularity and vessel permeability. The former is modeled as cerebral blood volume (CBV) in DSC and plasma fractional volume (Vp) in DCE. The value of CBV is often normalized with respect to a reference tissue, as denoted by relative CBV (rCBV) or normalized CBV (nCBV). Among existing studies using DSC or DCE for IDH mutation status assessment, discrepancies between different studies were evident. A significantly higher rCBV in IDH-wildtype compared with IDH-mutant type gliomas in all histological grades was reported in [20], whereas rCBV between IDH-wildtype and IDH-mutant gliomas did not differ significantly in histological subtypes of astrocytomas and oligodendrogliomas in [21]. Tissue vascularity was found to be significantly higher in IDH-wildtype gliomas than in IDH-mutant gliomas using DSC in [23]. However, tissue microenvironment parameters showed no correlates with glioma IDH mutation status using DCE in [24, 25] or DSC in [25]. The apparent conflicting results could be due to difference in imaging protocol, patient cohort, or tracer kinetic models for analyzing the acquired contrast-enhanced imaging data. Existing studies mostly utilized conventional tracer kinetic models such as the Tofts or extended Tofts model [26, 27], which does not differentiate the intravascular transport of tracer molecular with respect to the exchange process of tracer molecular between intravascular and interstitial spaces. There has been progress in the development of more advanced techniques in analyzing DCE data, such as the conventional compartment model (CC) [28], the adiabatic approximated tissue homogeneity model (ATH) [29], and the distributed parameter model (DP) [30]. The aforementioned two transports were separately accounted in these models, where blood flow (F) is utilized to characterize the intravascular transport and permeability-surface area product (PS) to describe the exchange between intravascular and interstitial spaces. In comparison, these two transports are modeled using one parameter, transfer constant (Ktrans), in the Tofts or extended Tofts model. Interested readers could refer to [31, 32] for a review on the topic. So far, few studies have been carried out on the investigation of these advanced tracer kinetic models in glioma molecular subtype characterization. Because advanced tracer kinetic models provide more realistic description of tracer transport in tissue microenvironment, it is expected that the derived parameters could be more interpretable with respect to tumor tissue microenvironment. This study hypothesizes that IDH mutations reduce the enzymatic activity of the encoded protein [7], leading to change in tissue microenvironment, and parameters derived using advanced tracer kinetic models would be more closely associated with glioma molecular signatures. Using DP as example, this study attempts to explore its application to glioma IDH mutation differentiation. 2. Materials and Methods 2.1. Subjects This retrospective study was approved by the institutional review board. Sixty-one patients were included in this study between August 2017 and September 2019. All patients diagnosed with gliomas of grade II–IV according to the 2016 WHO guideline on brain tumor classification after craniotomy and tumor resection. Patients in the study did not have a history of previous surgery for brain tumor. Six patients were excluded due to inadequate MRI quality. A total of 55 patients (23 men, 32 women; age range, 25–72 years; mean age, 46.45 ± 10.23 years) were included in the study. There were 7 oligodendrogliomas (WHO grade II), 11 astrocytomas (WHO grade II), 2 anaplastic oligodendrogliomas (WHO grade III), 8 anaplastic astrocytomas (WHO grade III), and 27 glioblastomas (WHO grade IV). Molecular pathological findings of IDH were determined by Sanger sequencing for IDH hotspot mutations. There were 24 patients with IDH mutation. A representative case is given in Figure 1. (a)
Article
The peritumoral vasogenic edema (PVE) in brain tumors exhibits varied characteristics. Brain metastasis (BM) and meningioma barely have tumor cells in PVE, while Glioblastoma (GB) show tumor cell infiltration in most subjects. The purpose of this study was to investigate the PVE of these three pathologies using Radiomics features in FLAIR images, with the hypothesis that the tumor cells might influence textural variation. Ex‐vivo experimentation of Radiomics analysis of T1‐weighted images of the culture medium with and without suspended tumor cells was also attempted to infer the possible influence of increasing tumor cells on Radiomics features. This retrospective study involved MR images acquired using 3.0T MR machine from 83 patients having 48 GB, 21 BM and 14 Meningioma. The 93 Radiomics features were extracted from each subject’s PVE mask from 3 pathologies using T1‐DCE MRI. Statistically significant (<0.05, independent samples T‐test) features were considered. Features maps were also computed for qualitative investigation. The same was carried out for T1‐weighted cell line images but group comparison was carried using One‐way ANOVA. Further, a random forest (RF) based machine learning (ML) model was designed to classify the PVE of GB and BM. The texture‐based variation especially higher non‐uniformity values were observed in the PVE of GB. No significance was observed between BM and Meningioma PVE. In cell line images, the culture medium had higher non‐uniformity and considerably reduced with increasing cell densities in 4 features. RF model implemented with highly significant features provided improved AUC results. The possible infiltrative tumor cells in the PVE of the GB are likely influencing the texture values and are higher in comparison with BM PVE and may be of value in differentiation of solitary metastasis from GB. However, the robustness of the features needs to be investigated with larger cohort and across the scanners in future.
Article
Purpose This study aimed to explore the feasibility of transmembrane water exchange parameters detected by brain shutter speed (BSS) dynamic contrast enhanced (DCE)MRI, which is validated to be associated with aquaporin-4 expression, in distinguishing glioblastoma (GBM) from solitary brain metastasis (SBM). Methods 40 patients (mean age: 58.6 ± 11.7 years old, male/female: 23/17) with GBM and 48 patients (mean age: 61.7 ± 10.5 years old, male/female: 28/20) with SBM were enrolled in this observational study. BSS DCE-MRI was performed before operation. Intravascular water efflux rate constant (kbo) and intracellular water efflux rate constant (kio) within the peritumoral region and enhancing tumor were calculated from SS-DCE, respectively. The difference of these two parameters between GBM and SBM was explored. Immunohistochemical staining aquaporin-4 of was performed to validate its underlying biological mechanism. Results The kbo was found to be statistically different within both peritumoral region {SBM vs. HGG (s-1): 1.0[0.4,1.7] vs. 1.5[0.9,2.1], p=0.009} and enhanced tumor {SBM vs. HGG (s-1): 0.2[0.1,0.5] vs. 0.4[0.1,1.3], p=0.034}. Immunohistochemical analysis reveals the high perivascular aquaporin-4 expression in GBM may contribute the higher kbo value than that of SBM. Conclusions kbo derived from BSS DCE-MRI was an independent pathophysiological parameter for separating GBM from SBM, in which kbo might be associated with the perivascular aquaporin-4 expression.
Article
Objective To evaluate the effect of artery input function (AIF) derived from different arteries for pharmacokinetic modeling on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) parameters in the grading of gliomas. Methods 49 patients with pathologically confirmed gliomas were recruited and underwent DCE-MRI. A modified Tofts model with different AIFs derived from anterior cerebral artery (ACA), ipsilateral and contralateral middle cerebral artery (MCA) and posterior cerebral artery (PCA) was used to estimate quantitative parameters such as K trans (volume transfer constant) and V e (fractional extracellular-extravascular space volume) for distinguishing the low grade glioma from high grade glioma. The K trans and V e were compared between different arteries using Two Related Samples Tests (TRST) (i.e. Wilcoxon Signed Ranks Test). In addition, these parameters were compared between the low and high grades as well as between the grade II and III using the Mann-Whitney U-test. A p-value of less than 0.05 was regarded as statistically significant. Results All the patients completed the DCE-MRI successfully. Sharp wash-in and wash-out phases were observed in all AIFs derived from the different arteries. The quantitative parameters (K trans and V e ) calculated from PCA were significant higher than those from ACA and MCA for low and high grades, respectively (p < 0.05). Despite the differences of quantitative parameters derived from ACA, MCA and PCA, the K trans and V e from any AIFs could distinguish between low and high grade, however, only K trans from any AIFs could distinguish grades II and III. There was no significant correlation between parameters and the distance from the artery, which the AIF was extracted, to the tumor. Conclusion Both quantitative parameters K trans and V e calculated using any AIF of ACA, MCA, and PCA can be used for distinguishing the low- from high-grade gliomas, however, only K trans can distinguish grades II and III. Advances in knowledge We sought to assess the effect of AIF on DCE-MRI for determining grades of gliomas. Both quantitative parameters K trans and V e calculated using any AIF of ACA, MCA, and PCA can be used for distinguishing the low- from high-grade gliomas.