Revolutionizing Glioblastoma Immunotherapy Conquering Transport and Biological Challenges, Innovating Combinatorial Approaches for Unprecedented Treatment Success
Glioblastomas are the most common primary brain cancer and present many challenges in treatment, with the current standard-of-care treatments yielding a median survival rate of less than 15 months. While immunotherapy against cancer has been very effective in some cancers, its application in glioblastoma has been limited so far. The following review touches upon some of the critical challenges associated with successful immunotherapy in glioblastoma, covering transport-related obstacles presented by the blood-brain barrier, biological complexity within the central nervous system, and the interplay between glioblastoma and immune cells. Ongoing clinical trials testing the efficacy of different immunotherapeutic strategies, including immune checkpoint blockade, vaccination, and adoptive cell transfer, are discussed. These strategies are inherently challenged by the low immunogenicity of glioblastoma, the unique immune-protective mechanisms of the immune system within the CNS, and the predominant features of the immune-suppressive tumor microenvironment. Current therapeutic modalities reviewed include surgical resection, radiation therapy, and temozolomide-based chemotherapy, with discussions on new forms of approaches to enhance immune activation: vaccines, oncolytic viruses, and adoptive cell therapies such as CAR T cells and NK cells. The perpetual problem of resistance to immunotherapy underlines the need for combination strategies and precise testing within advanced in-vitro and animal tumor models, considering the large variability in glioblastomas.
Ostrom
Q.T.
Gittleman
H.
Liao
P.
Vecchione-Koval
T.
Wolinsky
Y.
Kruchko
C.
Barnholtz-Sloan
J.S.
CBTRUS Statistical Report: Primary brain and other central nervous system tumors diagnosed in the United States in 2010–2014.
Neuro-oncol.2017
19
Suppl. 5
v1
v88
10.1093/neuonc/nox158
29117289
Roberts
Z.J.
Better
M.
Bot
A.
Roberts
M.R.
Ribas
A.
Axicabtagene ciloleucel, a first-in-class CAR T cell therapy for aggressive NHL.
Leuk. Lymphoma2018
59
8
1785
1796
10.1080/10428194.2017.1387905
29058502
Rosic
G.
Selakovic
D.
Omarova
S.
Cancer signaling, cell/gene therapy, diagnosis and role of nanobiomaterials.
Advances in Biology & Earth Sciences2024
9
Special Issue
11
34
10.62476/abes9s11
Xu
J.
Biosynthesis, characterization, and investigation of antimicrobial and cytotoxic activities of silver nanoparticles using Solanum tuberosum peel aqueous extract.
Heliyon2023
9
8
İpek
P.
Green synthesis and evaluation of antipathogenic, antioxidant, and anticholinesterase activities of gold nanoparticles (Au NPs) from Allium cepa L. peel aqueous extract.
Biomass Conv. Bioref.2024
14
9
10661
10670
Sarkaria
J.N.
Hu
L.S.
Parney
I.F.
Pafundi
D.H.
Brinkmann
D.H.
Laack
N.N.
Giannini
C.
Burns
T.C.
Kizilbash
S.H.
Laramy
J.K.
Swanson
K.R.
Kaufmann
T.J.
Brown
P.D.
Agar
N.Y.R.
Galanis
E.
Buckner
J.C.
Elmquist
W.F.
Is the blood–brain barrier really disrupted in all glioblastomas? A critical assessment of existing clinical data.
Neuro-oncol.2018
20
2
184
191
10.1093/neuonc/nox175
29016900
Hashizume
H.
Baluk
P.
Morikawa
S.
McLean
J.W.
Thurston
G.
Roberge
S.
Jain
R.K.
McDonald
D.M.
Openings between defective endothelial cells explain tumor vessel leakiness.
Am. J. Pathol.2000
156
4
1363
1380
10.1016/S0002‑9440(10)65006‑7
10751361
Peer
D.
Karp
J.M.
Hong
S.
Farokhzad
O.C.
Margalit
R.
Langer
R.
Nanocarriers as an emerging platform for cancer therapy.
Nat. Nanotechnol.2007
2
12
751
760
10.1038/nnano.2007.387
18654426
Danhier
F.
To exploit the tumor microenvironment: Since the EPR effect fails in the clinic, what is the future of nanomedicine?
J. Control. Release2016
244
Pt A
108
121
10.1016/j.jconrel.2016.11.015
27871992
Hansen
A.E.
Petersen
A.L.
Henriksen
J.R.
Boerresen
B.
Rasmussen
P.
Elema
D.R.
RosenschöldPMa, Kristensen AT, Kjær A, Andresen TL. Positron emission tomography based elucidation of the enhanced permeability and retention efect in dogs with cancer using copper-64 liposomes.
ACS Nano2015
9
6985
6995
10.1021/acsnano.5b01324
26022907
Sindhwani
S.
Syed
A.M.
Ngai
J.
Kingston
B.R.
Maiorino
L.
Rothschild
J.
MacMillan
P.
Zhang
Y.
Rajesh
N.U.
Hoang
T.
Wu
J.L.Y.
Wilhelm
S.
Zilman
A.
Gadde
S.
Sulaiman
A.
Ouyang
B.
Lin
Z.
Wang
L.
Egeblad
M.
Chan
W.C.W.
The entry of nanoparticles into solid tumours.
Nat. Mater.2020
19
5
566
575
10.1038/s41563‑019‑0566‑2
31932672
Hodi
F.S.
O’Day
S.J.
McDermott
D.F.
Weber
R.W.
Sosman
J.A.
Haanen
J.B.
Gonzalez
R.
Robert
C.
Schadendorf
D.
Hassel
J.C.
Akerley
W.
van den Eertwegh
A.J.M.
Lutzky
J.
Lorigan
P.
Vaubel
J.M.
Linette
G.P.
Hogg
D.
Ottensmeier
C.H.
Lebbé
C.
Peschel
C.
Quirt
I.
Clark
J.I.
Wolchok
J.D.
Weber
J.S.
Tian
J.
Yellin
M.J.
Nichol
G.M.
Hoos
A.
Urba
W.J.
Improved survival with ipilimumab in patients with metastatic melanoma.
N. Engl. J. Med.2010
363
8
711
723
10.1056/NEJMoa1003466
20525992
Subhan
M.A.
Parveen
F.
Filipczak
N.
Yalamarty
S.S.K.
Torchilin
V.P.
Approaches to improve EPR-based drug delivery for cancer therapy and diagnosis.
J. Pers. Med.2023
13
3
389
10.3390/jpm13030389
36983571
Tian
Y.
Cheng
T.
Sun
F.
Zhou
Y.
Yuan
C.
Guo
Z.
Wang
Z.
Effect of biophysical properties of tumor extracellular matrix on intratumoral fate of nanoparticles: Implications on the design of nanomedicine.
Adv. Colloid Interface Sci.2024
326
103124
10.1016/j.cis.2024.103124
38461766
Tran
V.L.
Lux
F.
Tournier
N.
Jego
B.
Maître
X.
Anisorac
M.
Comtat
C.
Jan
S.
Selmeczi
K.
Evans
M.J.
Tillement
O.
Kuhnast
B.
Truillet
C.
Quantitative tissue pharmacokinetics and EPR effect of AGuIX nanoparticles: a multimodal imaging study in an orthotopic glioblastoma rat model and healthy macaque.
Adv. Healthc. Mater.2021
10
16
2100656
10.1002/adhm.202100656
34212539
Rutledge
W.C.
Kong
J.
Gao
J.
Gutman
D.A.
Cooper
L.A.D.
Appin
C.
Park
Y.
Scarpace
L.
Mikkelsen
T.
Cohen
M.L.
Aldape
K.D.
McLendon
R.E.
Lehman
N.L.
Miller
C.R.
Schniederjan
M.J.
Brennan
C.W.
Saltz
J.H.
Moreno
C.S.
Brat
D.J.
Tumor-infiltrating lymphocytes in glioblastoma are associated with specific genomic alterations and related to transcriptional class.
Clin. Cancer Res.2013
19
18
4951
4960
10.1158/1078‑0432.CCR‑13‑0551
23864165
Yeung
J.T.
Hamilton
R.L.
Ohnishi
K.
Ikeura
M.
Potter
D.M.
Nikiforova
M.N.
Ferrone
S.
Jakacki
R.I.
Pollack
I.F.
Okada
H.
LOH in the HLA class I region at 6p21 is associated with shorter survival in newly diagnosed adult glioblastoma.
Clin. Cancer Res.2013
19
7
1816
1826
10.1158/1078‑0432.CCR‑12‑2861
23401227
Grossman
S.A.
Ye
X.
Lesser
G.
Sloan
A.
Carraway
H.
Desideri
S.
Piantadosi
S.
Consortium
N.C.
Immunosuppression in patients with high-grade gliomas treated with radiation and temozolomide.
Clin. Cancer Res.2011
17
16
5473
5480
10.1158/1078‑0432.CCR‑11‑0774
21737504
Mazor
G.
Levin
L.
Picard
D.
Ahmadov
U.
Carén
H.
Borkhardt
A.
Reifenberger
G.
Leprivier
G.
Remke
M.
Rotblat
B.
The lncRNA TP73-AS1 is linked to aggressiveness in glioblastoma and promotes temozolomide resistance in glioblastoma cancer stem cells.
Cell Death Dis.2019
10
3
246
10.1038/s41419‑019‑1477‑5
30867410
Orzan
F.
De Bacco
F.
Crisafulli
G.
Pellegatta
S.
Mussolin
B.
Siravegna
G.
D’Ambrosio
A.
Comoglio
P.M.
Finocchiaro
G.
Boccaccio
C.
Genetic evolution of glioblastoma stem-like cells from primary to recurrent tumor.
Stem Cells2017
35
11
2218
2228
10.1002/stem.2703
28895245
Young
R.M.
Jamshidi
A.
Davis
G.
Sherman
J.H.
Current trends in the surgical management and treatment of adult glioblastoma.
Ann. Transl. Med.2015
3
9
121
26207249
Brown
T.J.
Brennan
M.C.
Li
M.
Church
E.W.
Brandmeir
N.J.
Rakszawski
K.L.
Patel
A.S.
Rizk
E.B.
Suki
D.
Sawaya
R.
Glantz
M.
Association of the Extent of Resection With Survival in Glioblastoma.
JAMA Oncol.2016
2
11
1460
1469
10.1001/jamaoncol.2016.1373
27310651
Han
Q.
Liang
H.
Cheng
P.
Yang
H.
Zhao
P.
Gross Total vs. Subtotal Resection on Survival Outcomes in Elderly Patients With High-Grade Glioma: A Systematic Review and Meta-Analysis.
Front. Oncol.2020
10
151
10.3389/fonc.2020.00151
32257941
Cabrera
A.R.
Kirkpatrick
J.P.
Fiveash
J.B.
Shih
H.A.
Koay
E.J.
Lutz
S.
Petit
J.
Chao
S.T.
Brown
P.D.
Vogelbaum
M.
Reardon
D.A.
Chakravarti
A.
Wen
P.Y.
Chang
E.
Radiation therapy for glioblastoma: Executive summary of an American Society for Radiation Oncology Evidence-Based Clinical Practice Guideline.
Pract. Radiat. Oncol.2016
6
4
217
225
10.1016/j.prro.2016.03.007
27211230
Nabors
L.B.
Portnow
J.
Ammirati
M.
Baehring
J.
Brem
H.
Butowski
N.
Fenstermaker
R.A.
Forsyth
P.
Hattangadi-Gluth
J.
Holdhoff
M.
Howard
S.
Junck
L.
Kaley
T.
Kumthekar
P.
Loeffler
J.S.
Moots
P.L.
Mrugala
M.M.
Nagpal
S.
Pandey
M.
Parney
I.
Peters
K.
Puduvalli
V.K.
Ragsdale
J.
III
Rockhill
J.
Rogers
L.
Rusthoven
C.
Shonka
N.
Shrieve
D.C.
Sills
A.K.
Jr
Swinnen
L.J.
Tsien
C.
Weiss
S.
Wen
P.Y.
Willmarth
N.
Bergman
M.A.
Engh
A.
NCCN guidelines insights: Central nervous system cancers, version 1.
J. Natl. Compr. Canc. Netw.2017
15
11
1331
1345
10.6004/jnccn.2017.0166
29118226
Reardon
D.A.
Brandes
A.A.
Omuro
A.
Mulholland
P.
Lim
M.
Wick
A.
Baehring
J.
Ahluwalia
M.S.
Roth
P.
Bähr
O.
Phuphanich
S.
Sepulveda
J.M.
De Souza
P.
Sahebjam
S.
Carleton
M.
Tatsuoka
K.
Taitt
C.
Zwirtes
R.
Sampson
J.
Weller
M.
Effect of nivolumab vs bevacizumab in patients with recurrent glioblastoma: The checkmate 143 phase 3 randomized clinical trial.
JAMA Oncol.2020
6
7
1003
1010
10.1001/jamaoncol.2020.1024
32437507
Ray
A.
Manjila
S.
Hdeib
A.M.
Radhakrishnan
A.
Nock
C.J.
Cohen
M.L.
Sloan
A.
Extracranial metastasis of gliobastoma: Three illustrative cases and current review of the molecular pathology and management strategies.
Mol. Clin. Oncol.2015
3
3
479
486
10.3892/mco.2015.494
26137254
Rosen
J.
Blau
T.
Grau
S.J.
Barbe
M.T.
Fink
G.R.
Galldiks
N.
Extracranial metastases of a cerebral glioblastoma: A case report and review of the literature.
Case Rep. Oncol.2018
11
2
591
600
10.1159/000492111
30283316
Rossi
J.
Giaccherini
L.
Cavallieri
F.
Napoli
M.
Moratti
C.
Froio
E.
Serra
S.
Fraternali
A.
Ghadirpour
R.
Cozzi
S.
Ciammella
P.
Iaccarino
C.
Pascarella
R.
Valzania
F.
Pisanello
A.
Extracranial metastases in secondary glioblastoma multiforme: a case report.
BMC Neurol.2020
20
1
382
10.1186/s12883‑020‑01959‑y
33087049
Weenink
B.
Draaisma
K.
Ooi
H.Z.
Kros
J.M.
Sillevis Smitt
P.A.E.
Debets
R.
French
P.J.
Low-grade glioma harbors few CD8 T cells, which is accompanied by decreased expression of chemo-attractants, not immunogenic antigens.
Sci. Rep.2019
9
1
14643
10.1038/s41598‑019‑51063‑6
31601888
Keskin
D.B.
Anandappa
A.J.
Sun
J.
Tirosh
I.
Mathewson
N.D.
Li
S.
Oliveira
G.
Giobbie-Hurder
A.
Felt
K.
Gjini
E.
Shukla
S.A.
Hu
Z.
Li
L.
Le
P.M.
Allesøe
R.L.
Richman
A.R.
Kowalczyk
M.S.
Abdelrahman
S.
Geduldig
J.E.
Charbonneau
S.
Pelton
K.
Iorgulescu
J.B.
Elagina
L.
Zhang
W.
Olive
O.
McCluskey
C.
Olsen
L.R.
Stevens
J.
Lane
W.J.
Salazar
A.M.
Daley
H.
Wen
P.Y.
Chiocca
E.A.
Harden
M.
Lennon
N.J.
Gabriel
S.
Getz
G.
Lander
E.S.
Regev
A.
Ritz
J.
Neuberg
D.
Rodig
S.J.
Ligon
K.L.
Suvà
M.L.
Wucherpfennig
K.W.
Hacohen
N.
Fritsch
E.F.
Livak
K.J.
Ott
P.A.
Wu
C.J.
Reardon
D.A.
Neoantigen vaccine generates intratumoral T cell responses in phase Ib glioblastoma trial.
Nature2019
565
7738
234
239
10.1038/s41586‑018‑0792‑9
30568305
Dutoit
V.
Migliorini
D.
Dietrich
P.Y.
Walker
P.R.
Immunotherapy of malignant tumors in the brain: How different from other sites?
Front. Oncol.2016
6
256
10.3389/fonc.2016.00256
28003994
Shraibman
B.
Barnea
E.
Kadosh
D.M.
Haimovich
Y.
Slobodin
G.
Rosner
I.
López-Larrea
C.
Hilf
N.
Kuttruff
S.
Song
C.
Britten
C.
Castle
J.
Kreiter
S.
Frenzel
K.
Tatagiba
M.
Tabatabai
G.
Dietrich
P.Y.
Dutoit
V.
Wick
W.
Platten
M.
Winkler
F.
von Deimling
A.
Kroep
J.
Sahuquillo
J.
Martinez-Ricarte
F.
Rodon
J.
Lassen
U.
Ottensmeier
C.
van der Burg
S.H.
Thor Straten
P.
Poulsen
H.S.
Ponsati
B.
Okada
H.
Rammensee
H.G.
Sahin
U.
Singh
H.
Admon
A.
Identification of tumor antigens among the hla peptidomes of glioblastoma tumors and plasma.
Mol. Cell. Proteomics2019
18
6
1255
1268
10.1074/mcp.RA119.001524
31154438
Facoetti
A.
Nano
R.
Zelini
P.
Morbini
P.
Benericetti
E.
Ceroni
M.
Campoli
M.
Ferrone
S.
Human leukocyte antigen and antigen processing machinery component defects in astrocytic tumors.
Clin. Cancer Res.2005
11
23
8304
8311
10.1158/1078‑0432.CCR‑04‑2588
16322289
Schumacher
T.
Bunse
L.
Pusch
S.
Sahm
F.
Wiestler
B.
Quandt
J.
Menn
O.
Osswald
M.
Oezen
I.
Ott
M.
Keil
M.
Balß
J.
Rauschenbach
K.
Grabowska
A.K.
Vogler
I.
Diekmann
J.
Trautwein
N.
Eichmüller
S.B.
Okun
J.
Stevanović
S.
Riemer
A.B.
Sahin
U.
Friese
M.A.
Beckhove
P.
von Deimling
A.
Wick
W.
Platten
M.
A vaccine targeting mutant IDH1 induces antitumour immunity.
Nature2014
512
7514
324
327
10.1038/nature13387
25043048
Yin
J.
Valin
K.L.
Dixon
M.L.
Leavenworth
J.W.
The role of microglia and macrophages in cns homeostasis, autoimmunity, and cancer.
J. Immunol. Res.2017
2017
1
12
10.1155/2017/5150678
29410971
Orrego
E.
Castaneda
C.A.
Castillo
M.
Bernabe
L.A.
Casavilca
S.
Chakravarti
A.
Meng
W.
Garcia-Corrochano
P.
Villa-Robles
M.R.
Zevallos
R.
Mejia
O.
Deza
P.
Belmar-Lopez
C.
Ojeda
L.
Distribution of tumor-infiltrating immune cells in glioblastoma.
CNS Oncol.2018
7
4
CNS21
10.2217/cns‑2017‑0037
30299157
Eckerdt
F.
Platanias
L.C.
Emerging role of glioma stem cells in mechanisms of therapy resistance.
Cancers (Basel)2023
15
13
3458
10.3390/cancers15133458
37444568
Gupta
A.
Dwivedi
T.
A simplified overview of World Health Organization classification update of central nervous system tumors 2016.
J. Neurosci. Rural Pract.2017
8
4
629
641
10.4103/jnrp.jnrp_168_17
29204027
Rayati
M.
Mansouri
V.
Ahmadbeigi
N.
Gene therapy in glioblastoma multiforme: Can it be a role changer?
Heliyon2024
10
5
e27087
10.1016/j.heliyon.2024.e27087
38439834
DeCordova
S.
Shastri
A.
Tsolaki
A.G.
Yasmin
H.
Klein
L.
Singh
S.K.
Kishore
U.
Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma.
Front. Immunol.2020
11
1402
10.3389/fimmu.2020.01402
32765498
Varn
F.S.
Wang
Y.
Mullins
D.W.
Fiering
S.
Cheng
C.
Systematic pan-cancer analysis reveals immune cell interactions in the tumor microenvironment.
Cancer Res.2017
77
6
1271
1282
10.1158/0008‑5472.CAN‑16‑2490
28126714
Yang
I.
Tihan
T.
Han
S.J.
Wrensch
M.R.
Wiencke
J.
Sughrue
M.E.
Parsa
A.T.
CD8+ T-cell infiltrate in newly diagnosed glioblastoma is associated with long-term survival.
J. Clin. Neurosci.2010
17
11
1381
1385
10.1016/j.jocn.2010.03.031
20727764
Mostafa
H.
Pala
A.
Högel
J.
Hlavac
M.
Dietrich
E.
Westhoff
M.A.
Nonnenmacher
L.
Burster
T.
Georgieff
M.
Wirtz
C.R.
Schneider
E.M.
Immune phenotypes predict survival in patients with glioblastoma multiforme.
J. Hematol. Oncol.2016
9
1
77
10.1186/s13045‑016‑0272‑3
27585656
Groblewska
M.
Litman-Zawadzka
A.
Mroczko
B.
The role of selected chemokines and their receptors in the development of gliomas.
Int. J. Mol. Sci.2020
21
10
3704
10.3390/ijms21103704
32456359
Zhang
J.
Caruso
F.P.
Sa
J.K.
Justesen
S.
Nam
D.H.
Sims
P.
Ceccarelli
M.
Lasorella
A.
Iavarone
A.
The combination of neoantigen quality and T lymphocyte infiltrates identifies glioblastomas with the longest survival.
Commun. Biol.2019
2
1
135
10.1038/s42003‑019‑0369‑7
31044160
Platten
M.
Ochs
K.
Lemke
D.
Opitz
C.
Wick
W.
Microenvironmental clues for glioma immunotherapy.
Curr. Neurol. Neurosci. Rep.2014
14
4
440
10.1007/s11910‑014‑0440‑1
24604058
Roy
L.O.
Poirier
M.B.
Fortin
D.
Transforming growth factor-beta and its implication in the malignancy of gliomas.
Target. Oncol.2015
10
1
1
14
10.1007/s11523‑014‑0308‑y
24590691
Frei
K.
Gramatzki
D.
Tritschler
I.
Schroeder
J.J.
Espinoza
L.
Rushing
E.J.
Weller
M.
Transforming growth factor-β pathway activity in glioblastoma.
Oncotarget2015
6
8
5963
5977
10.18632/oncotarget.3467
25849941
Hazrati
A.
Soudi
S.
Malekpour
K.
Mahmoudi
M.
Rahimi
A.
Hashemi
S.M.
Varma
R.S.
Immune cells-derived exosomes function as a double-edged sword: role in disease progression and their therapeutic applications.
Biomark. Res.2022
10
1
30
10.1186/s40364‑022‑00374‑4
35550636
Lohr
J.
Ratliff
T.
Huppertz
A.
Ge
Y.
Dictus
C.
Ahmadi
R.
Grau
S.
Hiraoka
N.
Eckstein
V.
Ecker
R.C.
Korff
T.
von Deimling
A.
Unterberg
A.
Beckhove
P.
Herold-Mende
C.
Effector T-cell infiltration positively impacts survival of glioblastoma patients and is impaired by tumor-derived TGF-β.
Clin. Cancer Res.2011
17
13
4296
4308
10.1158/1078‑0432.CCR‑10‑2557
21478334
Platten
M.
Weller
M.
Wick
W.
Shaping the glioma immune microenvironment through tryptophan metabolism.
CNS Oncol.2012
1
1
99
106
10.2217/cns.12.6
25054303
Piccirillo
C.A.
Letterio
J.J.
Thornton
A.M.
McHugh
R.S.
Mamura
M.
Mizuhara
H.
Shevach
E.M.
CD4(+)CD25(+) regulatory T cells can mediate suppressor function in the absence of transforming growth factor β1 production and responsiveness.
J. Exp. Med.2002
196
2
237
246
10.1084/jem.20020590
12119348
Lynes
J.P.
Nwankwo
A.K.
Sur
H.P.
Sanchez
V.E.
Sarpong
K.A.
Ariyo
O.I.
Dominah
G.A.
Nduom
E.K.
Biomarkers for immunotherapy for treatment of glioblastoma.
J. Immunother. Cancer2020
8
1
e000348
10.1136/jitc‑2019‑000348
32474411
Weller
M.
Butowski
N.
Tran
D.D.
Recht
L.D.
Lim
M.
Hirte
H.
Ashby
L.
Mechtler
L.
Goldlust
S.A.
Iwamoto
F.
Drappatz
J.
O’Rourke
D.M.
Wong
M.
Hamilton
M.G.
Finocchiaro
G.
Perry
J.
Wick
W.
Green
J.
He
Y.
Turner
C.D.
Yellin
M.J.
Keler
T.
Davis
T.A.
Stupp
R.
Sampson
J.H.
Butowski
N.
Campian
J.
Recht
L.
Lim
M.
Ashby
L.
Drappatz
J.
Hirte
H.
Iwamoto
F.
Mechtler
L.
Goldlust
S.
Becker
K.
Barnett
G.
Nicholas
G.
Desjardins
A.
Benkers
T.
Wagle
N.
Groves
M.
Kesari
S.
Horvath
Z.
Merrell
R.
Curry
R.
O’Rourke
J.
Schuster
D.
Wong
M.
Mrugala
M.
Jensen
R.
Trusheim
J.
Lesser
G.
Belanger
K.
Sloan
A.
Purow
B.
Fink
K.
Raizer
J.
Schulder
M.
Nair
S.
Peak
S.
Perry
J.
Brandes
A.
Weller
M.
Mohile
N.
Landolfi
J.
Olson
J.
Finocchiaro
G.
Jennens
R.
DeSouza
P.
Robinson
B.
Crittenden
M.
Shih
K.
Flowers
A.
Ong
S.
Connelly
J.
Hadjipanayis
C.
Giglio
P.
Mott
F.
Mathieu
D.
Lessard
N.
Sepulveda
S.J.
Lövey
J.
Wheeler
H.
Inglis
P-L.
Hardie
C.
Bota
D.
Lesniak
M.
Portnow
J.
Frankel
B.
Junck
L.
Thompson
R.
Berk
L.
McGhie
J.
Macdonald
D.
Saran
F.
Soffietti
R.
Blumenthal
D.
André de
S.B.C.M.
Nowak
A.
Singhal
N.
Hottinger
A.
Schmid
A.
Srkalovic
G.
Baskin
D.
Fadul
C.
Nabors
L.
LaRocca
R.
Villano
J.
Paleologos
N.
Kavan
P.
Pitz
M.
Thiessen
B.
Idbaih
A.
Frenel
J.S.
Domont
J.
Grauer
O.
Hau
P.
Marosi
C.
Sroubek
J.
Hovey
E.
Sridhar
P.S.
Cher
L.
Dunbar
E.
Coyle
T.
Raymond
J.
Barton
K.
Guarino
M.
Raval
S.
Stea
B.
Dietrich
J.
Hopkins
K.
Erridge
S.
Steinbach
J-P.
Pineda
L.E.
Balana
Q.C.
Sonia del
B.B.
Wenczl
M.
Molnár
K.
Hideghéty
K.
Lossos
A.
Myra van
L.
Levy
A.
Harrup
R.
Patterson
W.
Lwin
Z.
Sathornsumetee
S.
Lee
E-J.
Ho
J-T.
Emmons
S.
Duic
J.P.
Shao
S.
Ashamalla
H.
Weaver
M.
Lutzky
J.
Avgeropoulos
N.
Hanna
W.
Nadipuram
M.
Cecchi
G.
O’Donnell
R.
Pannullo
S.
Carney
J.
Hamilton
M.
MacNeil
M.
Beaney
R.
Fabbro
M.
Schnell
O.
Fietkau
R.
Stockhammer
G.
Malinova
B.
Odrazka
K.
Sames
M.
Miguel Gil
G.
Razis
E.
Lavrenkov
K.
Castro
G.
Ramirez
F.
Baldotto
C.
Viola
F.
Malheiros
S.
Lickliter
J.
Gauden
S.
Dechaphunkul
A.
Thaipisuttikul
I.
Thotathil
Z.
Ma
H-I.
Cheng
W-Y.
Chang
C-H.
Salas
F.
Dietrich
P-Y.
Mamot
C.
Nayak
L.
Nag
S.
Rindopepimut with temozolomide for patients with newly diagnosed, EGFRvIII-expressing glioblastoma (ACT IV): A randomised, double-blind, international phase 3 trial.
Lancet Oncol.2017
18
10
1373
1385
10.1016/S1470‑2045(17)30517‑X
28844499
Lim
M.
Xia
Y.
Bettegowda
C.
Weller
M.
Current state of immunotherapy for glioblastoma.
Nat. Rev. Clin. Oncol.2018
15
7
422
442
10.1038/s41571‑018‑0003‑5
29643471
Suryadevara
C.M.
Desai
R.
Abel
M.L.
Riccione
K.A.
Batich
K.A.
Shen
S.H.
Chongsathidkiet
P.
Gedeon
P.C.
Elsamadicy
A.A.
Snyder
D.J.
Herndon
J.E.
II
Healy
P.
Archer
G.E.
Choi
B.D.
Fecci
P.E.
Sampson
J.H.
Sanchez-Perez
L.
Temozolomide lymphodepletion enhances CAR abundance and correlates with antitumor efficacy against established glioblastoma.
OncoImmunology2018
7
6
e1434464
10.1080/2162402X.2018.1434464
29872570
Liau
L.M.
Ashkan
K.
Tran
D.D.
Campian
J.L.
Trusheim
J.E.
Cobbs
C.S.
Heth
J.A.
Salacz
M.
Taylor
S.
D’Andre
S.D.
First results on survival from a large Phase 3 clinical trial of an autologous dendritic cell vaccine in newly diagnosed glioblastoma.
J. Transl. Med.2018
16
1
Eoli
M.
Corbetta
C.
Anghileri
E.
Di Ianni
N.
Milani
M.
Cuccarini
V.
Musio
S.
Paterra
R.
Frigerio
S.
Nava
S.
Lisini
D.
Pessina
S.
Maddaloni
L.
Lombardi
R.
Tardini
M.
Ferroli
P.
DiMeco
F.
Bruzzone
M.G.
Antozzi
C.
Pollo
B.
Finocchiaro
G.
Pellegatta
S.
Expansion of effector and memory T cells is associated with increased survival in recurrent glioblastomas treated with dendritic cell immunotherapy.
Neurooncol. Adv.2019
1
1
vdz022
10.1093/noajnl/vdz022
32642658
Jiang
H.
Rivera-Molina
Y.
Gomez-Manzano
C.
Clise-Dwyer
K.
Bover
L.
Vence
L.M.
Yuan
Y.
Lang
F.F.
Toniatti
C.
Hossain
M.B.
Fueyo
J.
Oncolytic adenovirus and tumor-targeting immune modulatory therapy improve autologous cancer vaccination.
Cancer Res.2017
77
14
3894
3907
10.1158/0008‑5472.CAN‑17‑0468
28566332
Diefenbach
A.
Raulet
D.H.
The innate immune response to tumors and its role in the induction of T‐cell immunity.
Immunol. Rev.2002
188
1
9
21
10.1034/j.1600‑065X.2002.18802.x
12445277
O’Rourke
D.M.
Nasrallah
M.P.
Desai
A.
Melenhorst
J.J.
Mansfield
K.
Morrissette
J.J.D.
Martinez-Lage
M.
Brem
S.
Maloney
E.
Shen
A.
Isaacs
R.
Mohan
S.
Plesa
G.
Lacey
S.F.
Navenot
J.M.
Zheng
Z.
Levine
B.L.
Okada
H.
June
C.H.
Brogdon
J.L.
Maus
M.V.
A single dose of peripherally infused EGFRvIII-directed CAR T cells mediates antigen loss and induces adaptive resistance in patients with recurrent glioblastoma.
Sci. Transl. Med.2017
9
399
eaaa0984
10.1126/scitranslmed.aaa0984
28724573
Medikonda
R.
Dunn
G.
Rahman
M.
Fecci
P.
Lim
M.
A review of glioblastoma immunotherapy.
J. Neurooncol.2021
151
1
41
53
10.1007/s11060‑020‑03448‑1
32253714
Klein
E.
Hau
A.C.
Oudin
A.
Golebiewska
A.
Niclou
S.P.
Glioblastoma organoids: Pre-clinical applications and challenges in the context of immunotherapy.
Front. Oncol.2020
10
604121
10.3389/fonc.2020.604121
33364198
Lee
J.
Kotliarova
S.
Kotliarov
Y.
Li
A.
Su
Q.
Donin
N.M.
Pastorino
S.
Purow
B.W.
Christopher
N.
Zhang
W.
Park
J.K.
Fine
H.A.
Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines.
Cancer Cell2006
9
5
391
403
10.1016/j.ccr.2006.03.030
16697959
Kaur
K.
Topchyan
P.
Kozlowska
A.K.
Ohanian
N.
Chiang
J.
Maung
P.O.
Park
S.H.
Ko
M.W.
Fang
C.
Nishimura
I.
Jewett
A.
Super-charged NK cells inhibit growth and progression of stem-like/poorly differentiated oral tumors in vivo in humanized BLT mice; effect on tumor differentiation and response to chemotherapeutic drugs.
OncoImmunology2018
7
5
e1426518
10.1080/2162402X.2018.1426518
29721395
Merz
F.
Gaunitz
F.
Dehghani
F.
Renner
C.
Meixensberger
J.
Gutenberg
A.
Giese
A.
Schopow
K.
Hellwig
C.
Schäfer
M.
Bauer
M.
Stöcker
H.
Taucher-Scholz
G.
Durante
M.
Bechmann
I.
Organotypic slice cultures of human glioblastoma reveal different susceptibilities to treatments.
Neuro-oncol.2013
15
6
670
681
10.1093/neuonc/not003
23576601
Parker
J.J.
Lizarraga
M.
Waziri
A.
Foshay
K.M.
A human glioblastoma organotypic slice culture model for study of tumor cell migration and patient-specific effects of anti-invasive drugs.
J. Vis. Exp.2017
2017
125
53557
28784966
Hubert
C.G.
Rivera
M.
Spangler
L.C.
Wu
Q.
Mack
S.C.
Prager
B.C.
Couce
M.
McLendon
R.E.
Sloan
A.E.
Rich
J.N.
A three-dimensional organoid culture system derived from human glioblastomas recapitulates the hypoxic gradients and cancer stem cell heterogeneity of tumors found in vivo.Cancer Res.2016
76
8
2465
2477
10.1158/0008‑5472.CAN‑15‑2402
26896279
Jacob
F.
Salinas
R.D.
Zhang
D.Y.
Nguyen
P.T.T.
Schnoll
J.G.
Wong
S.Z.H.
Thokala
R.
Sheikh
S.
Saxena
D.
Prokop
S.
Liu
D.
Qian
X.
Petrov
D.
Lucas
T.
Chen
H.I.
Dorsey
J.F.
Christian
K.M.
Binder
Z.A.
Nasrallah
M.
Brem
S.
O’Rourke
D.M.
Ming
G.
Song
H.
A patient-derived glioblastoma organoid model and biobank recapitulates inter- and intra-tumoral heterogeneity.
Cell2020
180
1
188
204.e22
10.1016/j.cell.2019.11.036
31883794
Olson
B.
Li
Y.
Lin
Y.
Liu
E.T.
Patnaik
A.
Mouse models for cancer immunotherapy research.
Cancer Discov.2018
8
11
1358
1365
10.1158/2159‑8290.CD‑18‑0044
30309862
Zhao
Y.
Shuen
T.W.H.
Toh
T.B.
Chan
X.Y.
Liu
M.
Tan
S.Y.
Fan
Y.
Yang
H.
Lyer
S.G.
Bonney
G.K.
Loh
E.
Chang
K.T.E.
Tan
T.C.
Zhai
W.
Chan
J.K.Y.
Chow
E.K.H.
Chee
C.E.
Lee
G.H.
Dan
Y.Y.
Chow
P.K.H.
Toh
H.C.
Lim
S.G.
Chen
Q.
Development of a new patient-derived xenograft humanised mouse model to study human-specific tumour microenvironment and immunotherapy.
Gut2018
67
10
1845
1854
10.1136/gutjnl‑2017‑315201
29602780
Lin
S.
Huang
G.
Cheng
L.
Li
Z.
Xiao
Y.
Deng
Q.
Jiang
Y.
Li
B.
Lin
S.
Wang
S.
Wu
Q.
Yao
H.
Cao
S.
Li
Y.
Liu
P.
Wei
W.
Pei
D.
Yao
Y.
Wen
Z.
Zhang
X.
Wu
Y.
Zhang
Z.
Cui
S.
Sun
X.
Qian
X.
Li
P.
Establishment of peripheral blood mononuclear cell-derived humanized lung cancer mouse models for studying efficacy of PD-L1/PD-1 targeted immunotherapy.
MAbs2018
10
8
1301
1311
10.1080/19420862.2018.1518948
30204048
Bao
S.
Wu
Q.
McLendon
R.E.
Hao
Y.
Shi
Q.
Hjelmeland
A.B.
Dewhirst
M.W.
Bigner
D.D.
Rich
J.N.
Glioma stem cells promote radioresistance by preferential activation of the DNA damage response.
Nature2006
444
7120
756
760
10.1038/nature05236
17051156
Haddad
A.F.
Young
J.S.
Amara
D.
Berger
M.S.
Raleigh
D.R.
Aghi
M.K.
Butowski
N.A.
Mouse models of glioblastoma for the evaluation of novel therapeutic strategies.
Neurooncol. Adv.2021
3
1
vdab100
10.1093/noajnl/vdab100
34466804
Wo
J.Y.
Viswanathan
A.N.
Impact of radiotherapy on fertility, pregnancy, and neonatal outcomes in female cancer patients.
Int. J. Radiat. Oncol. Biol. Phys.2009
73
5
1304
1312
10.1016/j.ijrobp.2008.12.016
Le Rhun
E.
Preusser
M.
Roth
P.
Reardon
D.A.
van den Bent
M.
Wen
P.
Reifenberger
G.
Weller
M.
Molecular targeted therapy of glioblastoma.
Cancer Treat. Rev.2019
80
101896
10.1016/j.ctrv.2019.101896
31541850
Wang
T.
Shigdar
S.
Gantier
M.P.
Hou
Y.
Wang
L.
Li
Y.
Shamaileh
H.A.
Yin
W.
Zhou
S.F.
Zhao
X.
Duan
W.
Cancer stem cell targeted therapy: progress amid controversies.
Oncotarget2015
6
42
44191
44206
10.18632/oncotarget.6176
26496035
Shergalis
A.
Bankhead
A.
III
Luesakul
U.
Muangsin
N.
Neamati
N.
Current challenges and opportunities in treating glioblastoma.
Pharmacol. Rev.2018
70
3
412
445
10.1124/pr.117.014944
29669750
Belden
C.J.
Valdes
P.A.
Ran
C.
Pastel
D.A.
Harris
B.T.
Fadul
C.E.
Israel
M.A.
Paulsen
K.
Roberts
D.W.
Genetics of glioblastoma: a window into its imaging and histopathologic variability.
Radiographics2011
31
6
1717
1740
10.1148/rg.316115512
21997991
Schiffer
D.
Annovazzi
L.
Casalone
C.
Corona
C.
Mellai
M.
Glioblastoma: microenvironment and niche concept.
Cancers (Basel)2018
11
1
5
10.3390/cancers11010005
30577488
Razavi
S.M.
Lee
K.E.
Jin
B.E.
Aujla
P.S.
Gholamin
S.
Li
G.
Immune evasion strategies of glioblastoma.
Front. Surg.2016
3
11
10.3389/fsurg.2016.00011
26973839
Martínez Bedoya
D.
Dutoit
V.
Migliorini
D.
Allogeneic CAR T cells: an alternative to overcome challenges of CAR T cell therapy in glioblastoma.
Front. Immunol.2021
12
640082
10.3389/fimmu.2021.640082
33746981
Taylor
T.E.
Furnari
F.B.
Cavenee
W.K.
Targeting EGFR for treatment of glioblastoma: molecular basis to overcome resistance.
Curr. Cancer Drug Targets2012
12
3
197
209
10.2174/156800912799277557
22268382
Garnier
D.
Meehan
B.
Kislinger
T.
Daniel
P.
Sinha
A.
Abdulkarim
B.
Nakano
I.
Rak
J.
Divergent evolution of temozolomide resistance in glioblastoma stem cells is reflected in extracellular vesicles and coupled with radiosensitization.
Neuro-oncol.2018
20
2
236
248
10.1093/neuonc/nox142
29016925
Doucette
T.
Rao
G.
Rao
A.
Shen
L.
Aldape
K.
Wei
J.
Dziurzynski
K.
Gilbert
M.
Heimberger
A.B.
Immune heterogeneity of glioblastoma subtypes: extrapolation from the cancer genome atlas.
Cancer Immunol. Res.2013
1
2
112
122
10.1158/2326‑6066.CIR‑13‑0028
24409449
Wang
Q.
Hu
B.
Hu
X.
Kim
H.
Squatrito
M.
Scarpace
L.
deCarvalho
A.C.
Lyu
S.
Li
P.
Li
Y.
Barthel
F.
Cho
H.J.
Lin
Y.H.
Satani
N.
Martinez-Ledesma
E.
Zheng
S.
Chang
E.
Sauvé
C.E.G.
Olar
A.
Lan
Z.D.
Finocchiaro
G.
Phillips
J.J.
Berger
M.S.
Gabrusiewicz
K.R.
Wang
G.
Eskilsson
E.
Hu
J.
Mikkelsen
T.
DePinho
R.A.
Muller
F.
Heimberger
A.B.
Sulman
E.P.
Nam
D.H.
Verhaak
R.G.W.
Tumor evolution of glioma-intrinsic gene expression subtypes associates with immunological changes in the microenvironment.
Cancer Cell2017
32
1
42
56.e6
10.1016/j.ccell.2017.06.003
28697342
Tang
X.
Zhang
S.
Fu
R.
Zhang
L.
Huang
K.
Peng
H.
Dai
L.
Chen
Q.
Therapeutic prospects of mRNA-based gene therapy for glioblastoma.
Front. Oncol.2019
9
1208
10.3389/fonc.2019.01208
31781503
Chen
J.
Li
Y.
Yu
T.S.
McKay
R.M.
Burns
D.K.
Kernie
S.G.
Parada
L.F.
A restricted cell population propagates glioblastoma growth after chemotherapy.
Nature2012
488
7412
522
526
10.1038/nature11287
22854781
Oh
H.C.
Shim
J.K.
Park
J.
Lee
J.H.
Choi
R.J.
Kim
N.H.
Kim
H.S.
Moon
J.H.
Kim
E.H.
Chang
J.H.
Yook
J.I.
Kang
S.G.
Combined effects of niclosamide and temozolomide against human glioblastoma tumorspheres.
J. Cancer Res. Clin. Oncol.2020
146
11
2817
2828
10.1007/s00432‑020‑03330‑7
32712753
Xie
X.P.
Laks
D.R.
Sun
D.
Ganbold
M.
Wang
Z.
Pedraza
A.M.
Bale
T.
Tabar
V.
Brennan
C.
Zhou
X.
Parada
L.F.
Quiescent human glioblastoma cancer stem cells drive tumor initiation, expansion, and recurrence following chemotherapy.
Dev. Cell2022
57
1
32
46.e8
10.1016/j.devcel.2021.12.007
35016005
Revolutionizing Glioblastoma Immunotherapy Conquering Transport and Biological Challenges, Innovating Combinatorial Approaches for Unprecedented Treatment Success
Revolutionizing Glioblastoma Immunotherapy Conquering Transport and Biological Challenges, Innovating Combinatorial Approaches for Unprecedented Treatment Success