The highly aggressive nature of glioblastoma carries a dismal prognosis despite aggressive multimodal therapy. Alternative treatment regimens, such as immunotherapies, are known to intensify the inflammatory response in the treatment field. Follow-up imaging in these scenarios often mimics disease progression on conventional MRI, making accurate evaluation extremely challenging. To this end, revised criteria for assessment of treatment response in high-grade gliomas were successfully proposed by the RANO Working Group to distinguish pseudoprogression from true progression, with intrinsic constraints related to the postcontrast T1-weighted MRI sequence. To address these existing limitations, our group proposes a more objective and quantifiable “treatment agnostic” model, integrating into the RANO criteria advanced multimodal neuroimaging techniques, such as diffusion tensor imaging (DTI), dynamic susceptibility contrast-perfusion weighted imaging (DSC-PWI), dynamic contrast enhanced (DCE)-MRI, MR spectroscopy, and amino acid-based positron emission tomography (PET) imaging tracers, along with artificial intelligence (AI) tools (radiomics, radiogenomics, and radiopathomics) and molecular information to address this complex issue of treatment-related changes versus tumor progression in “real-time”, particularly in the early posttreatment window. Our perspective delineates the potential of incorporating multimodal neuroimaging techniques to improve consistency and automation for the assessment of early treatment response in neuro-oncology.

Translational Relevance

An unmet need in neuro-oncology is the difficulty to distinguish glioblastoma progression from pseudoprogression as conventional MR images are challenging to differentiate signal from “noise” after standard-of-care and novel/alternative therapies, which is critical for appropriate therapeutic decision-making and prognostication. For example, a recent phase III study on dendritic cell vaccine, reporting a positive response to vaccine, took more than 15 years to complete, hundreds of patients, hundreds of millions of dollars, and for individual patients, the main endpoint is survival. Therefore, there is an urgent need for real-time endpoints that could provide iterative, dynamic feedback to inform clinicians making therapeutic decisions for standard-of-care, and “go/stop” decisions on clinical trials. This perspective highlights the role of recent advances in neuroimaging to inform clinicians to maintain continuity, reduce the need for surgical intervention, and provide reassurance in patients with predominance of treatment effects, aka, “pseudoprogression.” Conversely, in patients with true tumor progression, early intervention with a second-line treatment can be instituted sooner than later, with alternative therapies or repeat surgery.

Glioblastoma is the most common primary malignant brain tumor in adults and carries a dismal prognosis. Despite maximal safe surgical resection followed by concurrent chemoradiotherapy (CCRT) using temozolomide and adjuvant temozolomide, the prognosis remains very poor, with median overall survival (OS) of 15 to 17 months (1). To improve outcomes, early and accurate diagnosis of postoperative tumor progression (TP) is crucial because it affects clinical decisions for therapeutic “early” intervention and determining overall prognosis. Pseudoprogression can confound therapeutic decision-making as these patients often have methylated O6-methylguanine-DNA-methyltransferase (MGMT) promoter, respond well to temozolomide, and have superior OS compared with patients with TP (2). These patients are closely monitored with short-interval follow-up MRI scans and continue on adjuvant temozolomide. Conversely, patients with TP often require repeat biopsy/surgical resection and/or switching to alternative therapies such as tumor treating fields (TTFields), chemowafers, antiangiogenic therapy, or immunotherapy (3).

Pseudoprogression (PsP), aka “treatment-related change”, is most commonly seen within 12 weeks after the completion of CCRT, characterized by geographic necrosis, reactive gliosis, and vascular hyalinization, but it may occur later in the course of the disease, up to six months after treatment (4). Both TP and PsP can exhibit new/increasing enhancement within the radiation field with progressive enlargement of T2/FLAIR signal abnormality. Importantly, these morphologic changes reflect impairment of the blood-brain barrier and are, therefore, nonspecific surrogate markers, and can be seen in both TP and PsP. To this end, revised criteria for assessment of treatment response in high-grade gliomas were proposed by the RANO working group (5, 6) to distinguish PsP from TP.

The modified RANO (mRANO; ref. 5) criteria, as the former RANO criteria (6), include measurement of enhancing lesions via 2D-biperpendicular diameter and use of percentage thresholds to define response and progression, both depending on serial follow-up MRI examinations. The updates of mRANO include the removal of qualitative nonenhancing tumor assessment requirements by T2-FLAIR abnormality and the use of the postradiation time point as the baseline for newly diagnosed glioblastoma response assessment. RANO/mRANO criteria have remarkably impacted the retrospective examination in clinical trials (7) and could also be adapted for routine clinical practice. Nevertheless, advanced MRI techniques, including diffusion tensor imaging (DTI), dynamic susceptibility contrast-perfusion weighted imaging (DSC-PWI), dynamic contrast enhanced (DCE)-MRI and magnetic resonance spectroscopy (MRS), widely used as a routine in clinical neuro-oncologic imaging in many major centers, can substantively support conventional MRI findings not only in “real-time” but also in a more objective quantitative manner.

According to some studies (8, 9) there is a reasonable agreement between bidimensional (2D) and volumetric (3D) measurements of contrast-enhancing tumors, advocating for 2D measurements as a more practical alternative. However, higher reader discordance using bidirectional measurements has also been quoted by several studies (10–12), particularly in high-grade gliomas (5). This fact could reflect the current challenges of standard MRI interpretation to monitor glioma growth, (i) most enhancing glioblastomas have an irregular morphology and/or ill-defined margins and can also present with multifocal lesions (13); (ii) PsP affects vascular permeability, increasing the area of enhancement within the radiation field, overestimating tumor volume (13); (iii) antiangiogenic agents, such as bevacizumab, promote a marked decrease in contrast enhancement but no significant difference in OS, known as pseudo response, overestimating response rate on the basis of the use of contrast enhancement as the principal instrument (14, 15). Collectively, the increased regional heterogeneity observed on follow-up imaging makes 2D as well as 3D measurements less specific, thereby warranting the exploitation of additional advanced neuroimaging techniques and artificial intelligence (AI) tools in a multiparametric approach.

Malignant gliomas exhibit heterogeneous biological phenotypes, with areas of both neovascularized (enhancing) and infiltrative (nonenhancing) components (16). Due to the subjective nature of the interpretation of T2-FLAIR, this sequence was removed from the mRANO criteria to examine nonenhancing tumor growth. However, it is well known that nonenhancing tumor progression results in dramatically different postprogression survival in patients with glioblastoma (17); nevertheless, T2-FLAIR has a limited role in differentiating inflammatory changes versus tumor infiltration, confounding the response assessment, thereby warranting association with advanced MRI metrics, such as DSC-PWI, DTI, and MRS.

The use of cutoffs is necessary to standardize measurement comparison among different centers, and RANO/mRANO criteria rely on thresholds to define response and progression (≥50% decrease for partial response; PR and > 25% increase for progressive disease; PD in the sum of product diameters). Ellingson and colleagues (5) reported relative arbitrariness of RANO thresholds, which are not optimized on the basis of scientific data to show the best correlation with survival benefit or time to treatment failure. Also, using thresholds based on percentage change is particularly biased for small tumors in which low absolute increases in tumor size are interpreted as a substantial percentage difference (9). Conversely, the explosion of PD is not escalated depending on the growth rate and changes over time. Although the definition is necessarily arbitrary for comparison purposes, most glioblastomas present with mixed treatment response (admixture of TP and PsP or regional heterogeneity with TP in one area and PsP in another); for instance, classifying patients with glioblastoma as PsP considering the overall change in size when some areas are in fact progressing result in missing the early turning point for therapeutic intervention.

mRANO criteria use the postradiation time point as the baseline scan and require a minimum period of 12 weeks after the completion of CCRT and a repeat scan after four weeks for confirmation of TP/PsP unless the site of PD is distant from the radiation field or there is pathologic evidence of TP (5). Noteworthy, immediate postsurgical residual volume is an important prognostic factor, reducing OS in glioblastoma, and MRI, mainly within 72 hours postsurgery, is warranted to reduce confusion with postoperative reactive enhancement. mRANO criteria primarily rely on highly unpredictable, transient radiographic changes that often accompany the initial chemoradiation phase to use the postradiation MRI as the baseline scan. However, PsP can be present beyond 12 weeks (up to six months), and these first 12 weeks are a critical phase in which at least 50% of patients experiencing radiographic changes have true progression and are prevented from receiving alternative therapies for recurrent disease, thereby adversely affecting eventual outcomes (18).

The time for intervention can be even longer in patients with glioblastoma undergoing immunotherapy, assessed by the iRANO criteria. According to iRANO criteria, patients presenting with a lesion at the site of the original tumor, with a probable diagnosis of TP, or even showing new lesions at distant locations within the first six months of the onset of immunotherapy but clinically stable should continue with current immunotherapy. Furthermore, per iRANO, confirmation of radiographic progression can be performed at 12 weeks follow-up MRI instead of four weeks (19), a relative drawback, considering that patients with GBM have a low median OS and may be potentially receiving noneffective immunotherapy. In addition, trusting the patient's symptoms for changes in clinical management, while generally helpful, can also be misleading, as patients with a smaller TP or TP in nondominant locations will likely be relatively asymptomatic, preventing early institution of appropriate therapy, which may be surgical resection before too late (i.e., tumor spreading across corpus callosum or involving more eloquent regions and thereby becoming unresectable). Therefore, there is a pressing need to develop robust and quantitative imaging biomarkers which are “therapy agnostic” for reliably assessing “real-time” tumor dynamics and redefining the treatment response in patients with glioblastoma.

Noteworthy, utilizing physiologic and metabolic imaging parameters have been investigated to accurately determine treatment outcomes and define clinically beneficial endpoints, including neurologic and immunologic functions. In gliomas, cellular density and tumor grade are directly related to the degree of water restriction on diffusion-weighted imaging (DWI) with low apparent diffusion coefficient (ADC) values. In addition, ADC has shown value in patients treated with antiangiogenic agents and immunotherapy (20, 21). Specifically, minimum ADC values from enhancing areas could differentiate between inflammation and progressive tumor in patients treated with dendritic cell vaccine (22). The pathologic characteristics of tumor vasculature assessed by DSC–PWI can also help to discriminate between TP and PsP. In TP, there is a marked increase in angiogenesis that leads to an increase in relative cerebral blood volume (rCBV). In contrast, PsP is characterized by an increased inflammatory response, local accumulation of edema, and abnormal vascular permeability leading to decreased rCBV (23, 24).

In GBMs, spatial and temporal intratumoral heterogeneity can result in a mismatch in the findings from diverse neuroimaging parameters. Therefore, the use of a single imaging technique or parameter may not always be reliable for evaluating treatment response. Integrating the unique strengths of different imaging techniques, such as DTI and DSC–PWI, in a multiparametric approach showed a precise assessment of treatment response (25). A meta-analysis (26) showed the potential surrogate endpoint of multiparametric MRI for the assessment of early-treatment response in newly diagnosed glioblastoma treated with CCRT, with pooled sensitivity and specificity of 84% and 95%, respectively. In addition, the multiparametric analysis could accurately evaluate treatment response to immunotherapies, such as immune checkpoint inhibitors (27), EGFR variant III (EGFRvIII) targeted CAR T (28), and IL4 receptor–targeted immunotherapy (29) in patients with recurrent glioblastoma.

MRS can also add utility to the response assessment of nonenhancing tumors, differentiation of TP versus PsP, and genetic profiling of gliomas. Elevated lipid and low choline (Cho)/N- acetyl aspartate (NAA) ratios have been reported in association with PsP (2). Whole-brain echo-planar spectroscopic imaging (EPSI) following standard therapy also showed higher Cho/Creatine (Cr) and Cho/NAA ratios in patients with TP compared with PsP. EPSI can detect “invisible” tumor cells occult in conventional MRI sequences, which is useful for planning the course of treatment for these patients, such as optimizing the extent of resection and the dose and target area of radiation (2, 30). Several amino acid(aa)-based PET imaging tracers have emerged as alternative candidates for metabolic imaging of brain tumors. These tracers are characterized by high tumor-to-brain contrast based on their relatively high specificity for neoplastic cells and low accumulation in normal brain tissues (31).

Despite promising findings, metabolic and physiologic imaging techniques are associated with certain shortcomings. It is well known that even minor differences in hardware components or sequence parameters may result in significant changes in image contrast and signal intensity values, hindering the interpretation of imaging results from different treatment centers. Therefore, widespread adoption of these imaging techniques into routine clinical workflow requires standardization, harmonization of data acquisition, and processing protocols, along with the application of well-defined quality assessment/control procedures. Fortunately, consensus guidelines have been proposed to implement diffusion, perfusion, spectroscopy, and PET imaging techniques across different clinical sites (32–35). Additional improvements in this field require data sharing and large multicentric validation studies.

AI is an emerging translational field that holds promise to improve the precision of diagnostic and therapeutic methods. Radiomic, radiogenomic, and radiopathomic tools provide a means of noninvasive sampling of tumor microenvironments, allowing for a dynamic and comprehensive evaluation of regionally heterogeneous brain tumors (36–38). Although using machine learning–based models in distinguishing TP from PsP, some studies have demonstrated that multiparametric (anatomic MRI, ADC, and CBV) radiomics model perform significantly better than a monoparametric radiomics model using either conventional MRI, DWI, or PWI-derived parameters (39). These findings further emphasize the importance of applying multi-parametric approach even if we use more sophisticated machine learning–based models in comparison to traditionally used logistic regression models.

However, AI methods have been challenged by insufficient training, heterogeneity of imaging protocols across hospitals, and lack of generalization to new patient data. These challenges prompted the development of the ReSPOND (Radiomics Signatures for PrecisiON Diagnostics) consortium for glioblastoma [10 institutions, 3 continents, more than 3,300 de novo patients with GBM, and datasets from The Cancer Imaging Archive (TCIA)] to further develop and test AI-based biomarkers by moving from single-institution studies to generalized, well-validated predictive biomarkers (40). Notable, AI-based concepts are in continuous development but not fully integrated into daily practice. We, at this time, still depend on the combination of humans and computers (augmented intelligence) to incorporate information from images, AI tools, and electronic medical records to enhance the stratification of patients into more precise therapeutic pathways enabling dynamic treatment monitoring in this era of personalized medicine (41).

Beyond histologic and radiomics diagnosis, there is increasing recognition of integrating an objective assessment of molecular information. The integration of molecular data, such as MGMT methylation status, EGFRvIII, and isocitrate dehydrogenase (IDH) mutation, can further improve response assessment. It is well recognized that the methylation status of MGMT and IDH mutated gliomas are independent favorable prognostic factors in patients with GBM, and MGMT methylated patients are more frequently associated with PsP (42, 43). In addition, EGFRvIII has been explored extensively for targeted therapies, more recently in CAR T-cell immunotherapy (44). In the era of radiogenomics, imaging signatures of tumor genomics can help in identifying the patients who benefit from certain targeted therapies. Specifying molecular properties of gliomas and predicting their changes over time can also optimize treatment surveillance.

In conclusion, there is an urgent need for a reproducible, quantitative, and objective model to accurately evaluate treatment-related temporal changes in “real-time”, particularly in the early posttreatment window, to improve consistency and reduce variability in assessing outcomes in neuro-oncology. The quantitative, multiparametric neuroimaging approach substantiating the well-established mRANO criteria holds promise to further improve accuracy and automation in the future (Fig. 1).

Figure 1.

Schematic of future AI-based neuro-oncologic imaging and clinical management workflow for response assessment. A block diagram showing the trends in structural, metabolic, and physiologic neuroimaging derived parameters that are usually observed in distinguishing TP from PsP in glioblastomas. aa, amino acid; ADC, apparent diffusion coefficient; Cho, choline; CL, coefficient of linear anisotropy; CP, coefficient of planar anisotropy; Cr, creatine; CS, coefficient of spherical anisotropy; DCE, dynamic contrast-enhanced; DSC, dynamic susceptibility contrast; DTI, diffusion tensor imaging; DWI, diffusion weighted imaging; FA, fractional anisotropy; FET, O-(2-[18F] fluoroethyl)-L-tyrosine; GBM, glioblastoma; Ktrans, volume transfer constant; MD, mean diffusivity; MRI, magnetic resonance imaging; NAA, N-acetylaspartate; PC-T1, post-contrast T1 weighted images; PET, positron emission tomography; PsP, pseudo-progression; rCBV, relative cerebral blood volume; SPD, sum of product diameters; TP, true progression; Ve, volume fraction of extravascular-extracellular space in tissues; Vp, volume fraction of plasma space in tissues.

Figure 1.

Schematic of future AI-based neuro-oncologic imaging and clinical management workflow for response assessment. A block diagram showing the trends in structural, metabolic, and physiologic neuroimaging derived parameters that are usually observed in distinguishing TP from PsP in glioblastomas. aa, amino acid; ADC, apparent diffusion coefficient; Cho, choline; CL, coefficient of linear anisotropy; CP, coefficient of planar anisotropy; Cr, creatine; CS, coefficient of spherical anisotropy; DCE, dynamic contrast-enhanced; DSC, dynamic susceptibility contrast; DTI, diffusion tensor imaging; DWI, diffusion weighted imaging; FA, fractional anisotropy; FET, O-(2-[18F] fluoroethyl)-L-tyrosine; GBM, glioblastoma; Ktrans, volume transfer constant; MD, mean diffusivity; MRI, magnetic resonance imaging; NAA, N-acetylaspartate; PC-T1, post-contrast T1 weighted images; PET, positron emission tomography; PsP, pseudo-progression; rCBV, relative cerebral blood volume; SPD, sum of product diameters; TP, true progression; Ve, volume fraction of extravascular-extracellular space in tissues; Vp, volume fraction of plasma space in tissues.

Close modal

No disclosures were reported.

1.
Poon
MTC
,
Sudlow
CLM
,
Figueroa
JD
,
Brennan
PM
.
Longer-term (≥2 years) survival in patients with glioblastoma in population-based studies pre- and post-2005: a systematic review and meta-analysis
.
Sci Rep
2020;
10
:
11622
.
2.
Chawla
S
,
Bukhari
S
,
Afridi
OM
,
Wang
S
,
Yadav
SK
,
Akbari
H
, et al
.
Metabolic and physiologic magnetic resonance imaging in distinguishing true progression from pseudoprogression in patients with glioblastoma
.
NMR Biomed
2022
;
35
:
e4719
.
3.
Zhang
H
,
Wang
R
,
Yu
Y
,
Liu
J
,
Luo
T
,
Fan
F
.
Glioblastoma treatment modalities besides surgery
.
J Cancer
2019
;
10
:
4793
806
.
4.
Lee
J
,
Wang
N
,
Turk
S
,
Mohammed
S
,
Lobo
R
,
Kim
J
, et al
.
Discriminating pseudoprogression and true progression in diffuse infiltrating glioma using multi-parametric MRI data through deep learning
.
Sci Rep
2020
;
10
:
20331
.
5.
Ellingson
BM
,
Wen
PY
,
Cloughesy
TF
.
Modified criteria for radiographic response assessment in glioblastoma clinical trials
.
Neurotherapeutics
2017
;
14
:
307
20
.
6.
Wen
PY
,
Macdonald
DR
,
Reardon
DA
,
Cloughesy
TF
,
Sorensen
AG
,
Galanis
E
, et al
.
Updated response assessment criteria for high-grade gliomas: response assessment in neuro-oncology working group
.
J Clin Oncol
2010
;
28
:
1963
72
.
7.
Chukwueke
UN
,
Wen
PY
.
Use of the Response Assessment in Neuro-Oncology (RANO) criteria in clinical trials and clinical practice
.
CNS Oncol
2019
;
8
:
CNS28
.
8.
Galanis
E
,
Buckner
JC
,
Maurer
MJ
,
Sykora
R
,
Castillo
R
,
Ballman
KV
, et al
.
Validation of neuroradiologic response assessment in gliomas: measurement by RECIST, two-dimensional, computer-assisted tumor area, and computer-assisted tumor volume methods
.
Neuro Oncol
2006
;
8
:
156
65
.
9.
Ellingson
BM
,
Nguyen
HN
,
Lai
A
,
Nechifor
RE
,
Zaw
O
,
Pope
WB
, et al
.
Contrast-enhancing tumor growth dynamics of preoperative, treatment-naive human glioblastoma
.
Cancer
2016
;
122
:
1718
27
.
10.
Provenzale
JM
,
Ison
C
,
Delong
D
.
Bidimensional measurements in brain tumors: assessment of interobserver variability
.
AJR Am J Roentgenol
2009
;
193
:
W515
22
.
11.
Shah
GD
,
Kesari
S
,
Xu
R
,
Batchelor
TT
,
O'Neill
AM
,
Hochberg
FH
, et al
.
Comparison of linear and volumetric criteria in assessing tumor response in adult high-grade gliomas
.
Neuro Oncol
2006
;
8
:
38
46
.
12.
Thiesse
P
,
Ollivier
L
,
Di Stefano-Louineau
D
,
Négrier
S
,
Savary
J
,
Pignard
K
, et al
.
Response rate accuracy in oncology trials: reasons for interobserver variability. Groupe Français d'Immunothérapie of the Fédération Nationale des Centres de Lutte Contre le Cancer
.
J Clin Oncol
1997
;
15
:
3507
14
.
13.
Dempsey
MF
,
Condon
BR
,
Hadley
DM
.
Measurement of tumor “size” in recurrent malignant glioma: 1D, 2D, or 3D?
AJNR Am J Neuroradiol
2005
;
26
:
770
6
.
14.
Hygino da Cruz
LC
Jr
,
Rodriguez
I
,
Domingues
RC
,
Gasparetto
EL
,
Sorensen
AG
.
Pseudoprogression and pseudoresponse: imaging challenges in the assessment of posttreatment glioma
.
Am J Neuroradiol
2011
;
32
:
1978
85
.
15.
Norden
AD
,
Young
GS
,
Setayesh
K
,
Muzikansky
A
,
Klufas
R
,
Ross
GL
, et al
.
Bevacizumab for recurrent malignant gliomas: Efficacy, toxicity, and patterns of recurrence
.
Neurology
2008
;
70
:
779
87
.
16.
Chamberlain
MC
.
Radiographic patterns of relapse in glioblastoma
.
J Neurooncol
2011
;
101
:
319
23
.
17.
Nowosielski
M
,
Wiestler
B
,
Goebel
G
,
Hutterer
M
,
Schlemmer
HP
,
Stockhammer
G
, et al
.
Progression types after antiangiogenic therapy are related to outcome in recurrent glioblastoma
.
Neurology
2014
;
82
:
1684
92
.
18.
Ellingson
BM
,
Wen
PY
,
van den Bent
MJ
,
Cloughesy
TF
.
Pros and cons of current brain tumor imaging
.
Neuro-oncol
2014
;
16
:
page vii2–11
.
19.
Okada
H
,
Weller
M
,
Huang
R
,
Finocchiaro
G
,
Gilbert
MR
,
Wick
W
, et al
.
Immunotherapy response assessment in neuro-oncology: a report of the RANO working group
.
Lancet Oncol
2015
;
16
:
e534
42
.
20.
Zhang
M
,
Gulotta
B
,
Thomas
A
,
Kaley
T
,
Karimi
S
,
Gavrilovic
I
, et al
.
Large-volume low apparent diffusion coefficient lesions predict poor survival in bevacizumab-treated glioblastoma patients
.
Neuro-oncol
2016
;
18
:
735
43
.
21.
Wen
Q
,
Jalilian
L
,
Lupo
JM
,
Molinaro
AM
,
Chang
SM
,
Clarke
J
, et al
.
Comparison of ADC metrics and their association with outcome for patients with newly diagnosed glioblastoma being treated with radiation therapy, temozolomide, erlotinib and bevacizumab
.
J Neurooncol
2015
;
121
:
331
9
.
22.
Vrabec
M
,
Van Cauter
S
,
Himmelreich
U
,
Van Gool
SW
,
Sunaert
S
,
De Vleeschouwer
S
, et al
.
MR perfusion and diffusion imaging in the follow-up of recurrent glioblastoma treated with dendritic cell immunotherapy: a pilot study
.
Neuroradiology
2011
;
53
:
721
31
.
23.
Boxerman
JL
,
Ellingson
BM
,
Jeyapalan
S
,
Elinzano
H
,
Harris
RJ
,
Rogg
JM
, et al
.
Longitudinal DSC-MRI for distinguishing tumor recurrence from pseudoprogression in patients with a high-grade glioma
.
Am J Clin Oncol
2017
;
40
:
228
34
.
24.
Prager
AJ
,
Martinez
N
,
Beal
K
,
Omuro
A
,
Zhang
Z
,
Young
RJ
.
Diffusion and perfusion MRI to differentiate treatment-related changes including pseudoprogression from recurrent tumors in high-grade gliomas with histopathologic evidence
.
Am J Neuroradiol
2015
;
36
:
877
85
.
25.
Wang
S
,
Martinez-Lage
M
,
Sakai
Y
,
Chawla
S
,
Kim
SG
,
Alonso-Basanta
M
, et al
.
Differentiating tumor progression from pseudoprogression in patients with glioblastomas using diffusion tensor imaging and dynamic susceptibility contrast MRI
.
Am J Neuroradiol
2016
;
37
:
28
36
.
26.
Suh
CH
,
Kim
HS
,
Jung
SC
,
Choi
CG
,
Kim
SJ
.
Multiparametric MRI as a potential surrogate endpoint for decision-making in early treatment response following concurrent chemoradiotherapy in patients with newly diagnosed glioblastoma: a systematic review and meta-analysis
.
Eur Radiol
2018
;
28
:
2628
38
.
27.
Song
J
,
Kadaba
P
,
Kravitz
A
,
Hormigo
A
,
Friedman
J
,
Belani
P
, et al
.
Multiparametric MRI for early identification of therapeutic response in recurrent glioblastoma treated with immune checkpoint inhibitors
.
Neuro Oncol
2020
;
22
:
1658
66
.
28.
Wang
S
,
O'Rourke
DM
,
Chawla
S
,
Verma
G
,
Nasrallah
MP
,
Morrissette
JJD
, et al
.
Multiparametric magnetic resonance imaging in the assessment of anti-EGFRvIII chimeric antigen receptor T cell therapy in patients with recurrent glioblastoma
.
Br J Cancer
2019
;
120
:
54
6
.
29.
Mohan
S
,
Wang
S
,
Chawla
S
,
Abdullah
K
,
Desai
A
,
Maloney
E
, et al
.
Multiparametric MRI assessment of response to convection-enhanced intratumoral delivery of MDNA55, an interleukin-4 receptor targeted immunotherapy, for recurrent glioblastoma
.
Surg Neurol Int
2021
;
12
:
337
.
30.
Verma
G
,
Chawla
S
,
Mohan
S
,
Wang
S
,
Nasrallah
M
,
Sheriff
S
, et al
.
Three-dimensional echo planar spectroscopic imaging for differentiation of true progression from pseudoprogression in patients with glioblastoma
.
NMR Biomed
2019
;
32
:
e4042
.
31.
Drake
LR
,
Hillmer
AT
,
Cai
Z
.
Approaches to PET imaging of glioblastoma
.
Molecules
2020
;
25
:
568
.
32.
Ellingson
BM
,
Bendszus
M
,
Boxerman
J
,
Barboriak
D
,
Erickson
BJ
,
Smits
M
.
Consensus recommendations for a standardized brain tumor imaging protocol in clinical trials
.
Neuro-oncol
2015
;
17
:
1188
98
.
33.
Welker
K
,
Boxerman
J
,
Kalnin
A
,
Kaufmann
T
,
Shiroishi
M
,
Wintermark
M
, et al
.
ASFNR recommendations for clinical performance of MR dynamic susceptibility contrast perfusion imaging of the brain
.
AJNR Am J Neuroradiol
2015
;
36
:
E41
51
.
34.
Lin
A
,
Andronesi
O
,
Bogner
W
,
Choi
I-Y
,
Coello
E
,
Cudalbu
C
, et al
.
Minimum reporting standards for in vivo Magnetic Resonance Spectroscopy (MRSinMRS): experts’ consensus recommendations
.
NMR Biomed
2021
;
34
:
e4484
.
35.
Law
I
,
Albert
NL
,
Arbizu
J
,
Boellaard
R
,
Drzezga
A
,
Galldiks
N
, et al
.
Joint EANM/EANO/RANO practice guidelines/SNMMI procedure standards for imaging of gliomas using PET with radiolabelled amino acids and [F]FDG: version 1.0
.
Eur J Nucl Med Mol Imaging
2019
;
46
:
540
57
.
36.
Booth
TC
,
Grzeda
M
,
Chelliah
A
,
Roman
A
,
Al Busaidi
A
,
Dragos
C
, et al
.
Imaging biomarkers of glioblastoma treatment response: a systematic review and meta-analysis of recent machine learning studies
.
Front Oncol
2022
;
12
:
799662
.
37.
Akbari
H
,
Rathore
S
,
Bakas
S
,
Nasrallah
MP
,
Shukla
G
,
Mamourian
E
, et al
.
Histopathology-validated machine learning radiographic biomarker for noninvasive discrimination between true progression and pseudo-progression in glioblastoma
.
Cancer
2020
;
126
;
2625
36
.
38.
Fathi Kazerooni
A
,
Bagley
SJ
,
Akbari
H
,
Saxena
S
,
Bagheri
S
,
Guo
J
, et al
.
Applications of radiomics and radiogenomics in high-grade gliomas in the era of precision medicine
.
Cancers
2021
;
13
:
5921
.
39.
Kim
JY
,
Park
JE
,
Jo
Y
,
Shim
WH
,
Nam
SJ
,
Kim
JH
, et al
.
Incorporating diffusion- and perfusion-weighted MRI into a radiomics model improves diagnostic performance for pseudoprogression in glioblastoma patients
.
Neuro Oncol
2019
;
21
:
404
14
.
40.
Davatzikos
C
,
Barnholtz-Sloan
JS
,
Bakas
S
,
Colen
R
,
Mahajan
A
,
Quintero
CB
, et al
.
AI-based prognostic imaging biomarkers for precision neuro-oncology: the ReSPOND consortium
.
Neuro Oncol
2020
;
22
:
886
8
.
41.
Rudie
JD
,
Rauschecker
AM
,
Bryan
RN
,
Davatzikos
C
,
Mohan
S
.
Emerging applications of artificial intelligence in neuro-oncology
.
Radiology
2019
;
290
:
607
18
.
42.
Yang
P
,
Zhang
W
,
Wang
Y
,
Peng
X
,
Chen
B
,
Qiu
X
, et al
.
IDH mutation and MGMT promoter methylation in glioblastoma: results of a prospective registry
.
Oncotarget
2015
;
6
:
40896
906
.
43.
Li
M
,
Ren
X
,
Dong
G
,
Wang
J
,
Jiang
H
,
Yang
C
, et al
.
Distinguishing pseudoprogression from true early progression in isocitrate dehydrogenase wild-type glioblastoma by interrogating clinical, radiological, and molecular features
.
Front Oncol
2021
;
11
:
627325
.
44.
Chawla
S
,
Shehu
V
,
Gupta
PK
,
Nath
K
,
Poptani
H
.
Physiological imaging methods for evaluating response to immunotherapies in glioblastomas
.
Int J Mol Sci
2021
;
22
:
3867
.