Brain Immunology and Immunotherapy in Brain Tumours

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 14

RevIews

Brain immunology and immunotherapy


in brain tumours
John H. Sampson   1,2*, Michael D. Gunn   3, Peter E. Fecci   1,2,4 and David M. Ashley1
Abstract | Gliomas, the most common malignant primary brain tumours, remain universally lethal.
Yet, seminal discoveries in the past 5 years have clarified the anatomy , genetics and function
of the immune system within the central nervous system (CNS) and altered the paradigm for
successful immunotherapy. The impact of standard therapies on the response to immunotherapy
is now better understood, as well. This new knowledge has implications for a broad range of
tumours that develop within the CNS. Nevertheless, the requirements for successful therapy
remain effective delivery and target specificity , while the dramatic heterogeneity of malignant
gliomas at the genetic and immunological levels remains a profound challenge.

Tumour-​treating fields Malignant primary brain tumours are a leading cause of failure of a site to reject heterotopic tissue transplan-
(TTFields). External application cancer mortality in children and young adults, with few tation. In the case of the brain, these experiments are
of alternating electrical fields to therapeutic options. In adults, glioblastomas, the most habitually attributed to Peter Medawar in the 1940s
disrupt the multiplication of common primary brain tumours, remain uniformly (however, Medawar himself references the earlier work
tumour cells.
lethal, with a median survival of <21 months, despite of Shirai, Murphy and Tansley in his landmark 1948
Glial–lymphatic surgical resection, targeted radiation therapy, high-​dose paper)12. What is less well remembered of Medawar’s
(glymphatic) pathway chemotherapy and novel approaches such as tumour-​ studies is that while homologous skin grafting into
A recently characterized treating fields (TTFields)1–8. Despite the early identifica- brains failed to elicit immunity, skin homografts did
functional waste clearance
tion of genetic drivers of these tumours, they contain indeed succumb to rejection if their implantation fol-
pathway in the central nervous
system that connects the brain
relatively few coding mutations, and their intratumour lowed preliminary grafting of foreign tissue elsewhere
interstitium to the cerebrospinal heterogeneity has been shown to be extraordinary9,10. in the body. Thus, pre-​existing immune states could
fluid spaces via aquaporin In addition, unique features of these tumours and of extend to the brain to promote rejection, but such states
channels. the host organ thwart immunotherapeutic approaches. The could not be evoked de novo by central nervous sys-
immune system in the brain follows different principles tem (CNS) transplantation. Medawar attributed this
from the immune system elsewhere, whereby access to observed disparity to the perceived absence of a system
the tumours is limited by the blood–brain barrier (BBB), for lymphatic drainage, and therefore for afferent immu-
and the host is subjected to substantial endogenous and nity within the brain, a perception that has only recently
treatment-​induced immunosuppression. Thus, primary been disproved13–19.
1
The Preston Robert Tisch
Brain Tumor Center at Duke,
brain tumours are devastating diseases, and their unique In the many years since Medawar’s tissue-​grafting
Department of Neurosurgery, features are a topic of considerable importance. In this experi­ments, newer techniques have increasingly re­vealed
Duke University Medical Review, we discuss the immunological underpinnings the CNS immune privilege paradigm to be overstated.
Center, Durham, NC, USA. of brain metastases only briefly (Box 1) but, rather, focus Studies highlighting an existent afferent mechanism
2
Duke Brain Tumor on the specifics of the immune privilege of the brain, the for CNS participation in regional lymphatics debunked
Immunotherapy Program,
unique microenvironment of malignant glial tumours the isolationist idea of the brain as immunologically
Department of Neurosurgery,
Duke University Medical
and the limitations that the genetic underpinnings and silenced. For instance, routes of antigenic egress from
Center, Durham, NC, USA. standard therapies used place on the immunotherapeu- the brain to the deep cervical lymph nodes (via the
3
Department of Medicine, tic approaches and their success. We also summarize the arachnoid sheath of the olfactory nerves, through the
Duke University Medical existing immunotherapeutic approaches and biomarkers cribriform plate, and to the nasal mucosa) were uncov-
Center, Durham, NC, USA. of response that are currently under evaluation. ered beginning in the 1980s13–16. Identification of the
4
Duke Center for Brain and glial–lymphatic (glymphatic) pathway offered a mechanism
Spine Metastasis, Duke Brain tumour immunology is different for fluid and solute clearance from the brain, linking
University Medical Center,
Durham, NC, USA.
Immune privilege the parenchyma and interstitium to the cerebrospinal
The accessibility of the brain to the afferent and effer- fluid (CSF) spaces: CSF flows into the brain along arte-
*e-​mail: john.sampson@
duke.edu ent arms of the immune system, and thus to immuno- rial perivascular spaces and then translocates into the
https://doi.org/10.1038/ therapy, has been a matter of some debate for decades11. interstitium via aquaporin 4 (AQP4) water channels,
s41568-019-0224-7 Conceptually, the term ‘immune privilege’ refers to the before exiting along venous perivascular spaces20,21.

Nature Reviews | Cancer


Reviews

Glia limitans Since 2015, the discovery of functional lymphatic vessels Brain environmental cues influence the phenotype and
A thin barrier of astrocyte foot in the meninges has provided a direct drainage pathway activity of both microglia and recruited macrophages;
processes associated with the for CSF containing solute and immune cells from the however, the functions of these resident and recruited
parenchymal basal lamina brain into the cervical lymph nodes17–19. myeloid populations appear to differ in ways we do not
surrounding the brain and
spinal cord. The glia limitans
Given these provisions for an operative afferent yet fully understand36.
plays a crucial role in regulating immune system, many have proposed that the brain Despite the absence of the traditional manifestations
the movement of small is immunologically ‘distinct’ rather than ‘privileged’22 and protections of immune privilege, brain tumours
molecules and cells into the (Fig. 1). Such notions are bolstered by newer paradigms still possess the ability to prevent immunity and facili-
brain parenchyma.
of efferent CNS immunity, as well. Challenges for the tate their own modes of immune evasion. For instance,
Microglia efferent arm of CNS immunity have historically been a recent study has uncovered the capacity of intracra-
The resident self-​renewing ascribed to the BBB23. Formed by capillary tight junc- nial tumours, specifically, to sequester T cells in the
population of macrophages in tions and the astrocytic glia limitans24,25, the BBB serves bone marrow in a sphingosine 1 phosphate receptor 1
the central nervous system. as a structural barrier to the passive transit of molecules (S1PR1)-dependent fashion, preventing their surveil-
between the brain and the systemic circulation, permit- lance of the CNS and fostering antigenic ignorance37.
ting immune cell passage only at the level of the post-​ Thus, while our historical concept of CNS immune
capillary venules26. Ultimately, the CNS parenchyma privilege has perhaps become obsolete, the idiosyncra-
was believed to encourage only limited immunity, as sies of the brain environment continue to offer unique
direct antigenic challenges instigated a modest immune and formidable challenges to immune-​based platforms
infiltration when compared with systemic injections27. targeting those tumours harboured within38.
However, newer paradigms have documented T cell
entry and immunosurveillance within the brain26,28,29, Tumour microenvironment
disproving the notion that the BBB acts as a ‘hermetic Relative to other tumour types, CNS tumours dis-
seal’ to immune cell entry. Equally, damage incurred to play low numbers of tumour-​infiltrating lymphocytes
the BBB in the context of gliomas and other tumours (TILs) and other immune effector cell types39. This ‘cold
limits the restrictions normally proffered by the BBB30–32. tumour’ phenotype is associated with poor responses to
Another unique yet dynamic aspect of the brain immune stimulatory therapies such as immune check-
immune environment is its population of resident point blockade40. Even when T cell responses to CNS
myeloid cells33. At baseline, this population is com- tumours are induced through means such as vaccina-
posed almost entirely of microglia, the brain’s equivalent tion, antigen-​specific TIL numbers can remain relatively
of tissue-​resident macrophages. Microglia arise from low, and those cells that are present frequently display an
yolk sac myeloid progenitors that seed neural tissues exhausted phenotype41. The reduced quantity and lim-
during development and are maintained through repli­ ited activity of T cells in CNS tumours is largely due to
cation34. In the absence of inflammation, microglia the unique immune environment of the brain42. Owing
maintain their yolk sac origin throughout life, with no to its solid enclosure and the potential for damage from
contribution from bone marrow-​derived cells. After an increased intracranial pressure, unrestrained inflamma-
inflammatory stimulus, microglia undergo substan- tion in the brain poses a threat not seen in peripheral
tial phenotypic changes, while additional macrophage organs. For this reason, the CNS may have evolved to
populations are recruited from circulating monocytes35. be an environment in which both inflammatory and
adaptive immune responses are tightly regulated. This
Box 1 | Brain metastasis and immune checkpoint blockade regulation involves a variety of immunosuppressive
mechanisms at both the molecular and cellular levels43.
subversion of endogenous and manipulated immune responses, long a hallmark of
In response to inflammatory stimuli, including those
glioblastoma, is increasingly recognized among brain metastases, as well (reviewed
in Farber et al.230). The brain has evolved a variety of mechanisms aimed at curbing
derived from tumours, brain stromal cells produce
inflammatory responses that might otherwise be harmful, limiting both the access and remarkably high levels of the classic immunosuppres-
scope of T cell responses. Tumours of the intracranial compartment, in turn, are often sive cytokines transforming growth factor β (TGFβ) and
able to usurp such mechanisms to restrict the antitumour immune response. Similar interleukin-10 (IL-10), which counteract inflammatory
to glioblastoma, brain metastases frequently contain fewer T cell infiltrates than cytokines to maintain homeostasis44,45. Glioma cells
their peripherally located counterparts, and T cells that do successfully infiltrate are produce large amounts of indolamine 2,3-dioxygenase
subject to further suppressive influences geared at promoting such dysfunction as (IDO), which both stimulates the accumulation of
tolerance and exhaustion150,231. Tumour infiltrates within the brain are uniquely heavy in regu­latory T (Treg) cells and suppresses T cell activity by
microglia and monocytes, which may function ultimately to dampen the cell-​mediated depleting tryptophan from the microenvironment46,47.
immune response, a function employed by brain metastases232–234.
Both microglia and tumour-​infiltrating myeloid cells
While the systemic and local immunological consequences of CNS metastasis are not
as well characterized as in primary brain tumours, current data suggest contributions
produce high levels of arginase, which inhibits T cell
by many of the same microenvironmental immunosuppressive factors, including proliferation and function through the depletion of
regulatory T (Treg) cells154,235 and noteworthy expression of programmed cell death tissue arginine levels48. The brain appears to be particu-
protein 1 ligand 1 (PDL1)236. Numerous studies have documented T cell ineffectiveness larly susceptible to amino acid deprivation, as CSF argi-
that correlates with the expression of immune checkpoints150,160; however, unlike in nine and tryptophan levels depend on active transport
the case of glioblastoma, the available evidence suggests that immune checkpoint across the BBB and are sustained at only ~10% of the
blockade may proffer significant efficacy against intracranial metastases, particularly concentrations found in plasma49 (Fig. 2).
those from melanoma151. Nevertheless, future immunotherapeutic successes will The strategy of inhibiting specific immunosuppres-
depend on deepening our understanding of the interactions between metastatic sive factors is now being tested in patients with primary
tumours and immune populations within the brain specifically.
brain tumours, typically in combination with other

www.nature.com/nrc
Reviews

Microglia, macrophages, astrocytes and


can comprise up to 30% of the tumour mass59. In both
Neuron DCs are found within the CNS and may mouse models and humans, the vast majority of brain
have roles in immune surveillance TAMs appear to arise from circulating monocytes, with
DC a minor proportion (~15%) being microglia derived60–62.
Astrocyte TAMs are believed to promote tumour growth, and TAM
Functional numbers correlate with tumour grade63. TAM growth-​
lymphatic
Microglia promoting activity has been associated with the anti-​
vessels have inflammatory M2 macrophage phenotype64; however,
been observed
in the CNS Macrophage this likely represents an oversimplification, as a much
T cell
broader range of macrophage activation states occurs
CNS antigens Glioblastoma
drain through CSF microenvironment in vivo65.
A variety of approaches have been explored as means
to either reduce TAM numbers or reprogramme them to
be more inflammatory and immunogenic66. The cytokine
Activated T cells Lymph colony-​stimulating factor 1 (CSF1) promotes myeloid
• Cross the BBB node
• Patrol the CNS in an
cell differentiation and survival67. In mice, CSF1 recep-
unrestricted manner tor (CSF1R) inhibition with a small molecule markedly
• Return to systemic increased survival in a genetically engineered platelet-​
circulation
CNS antigens are circulated
derived growth factor β (PDGFβ)-driven model of gli-
to cervical lymph nodes oma68; however, no objective response was observed
in a phase II study (NCT01349036 (ref.69)) of a CSF1R
inhibitor in patients with recurrent glioblastoma70. The
Fig. 1 | Immune privilege in the brain. Historically , the central nervous system (CNS) chemotherapy agent trabectedin has been shown to
was considered to be isolated immunologically. Features that contributed to this
function, at least in part, by depleting the monocytes
understanding were the presence of tight junctions in the blood–brain barrier (BBB),
the absence of a classic lymphatic drainage system and empirical data showing limited
that serve as TAM precursors71. Other strategies, includ-
rejection of foreign tissues within the CNS. However, today the concept of immune ing the inhibition of CC-​chemokine ligand 2 (CCL2)–
privilege has been partially redefined. It is now clear that there are functional lymphatic CC-​chemokine receptor 2 (CCR2)-mediated TAM
vessels in the CNS, and that antigen-​presenting cells (APCs) of varied types exist accumulation with receptor antagonists72, the repro-
within the CNS, including microglia, macrophages, astrocytes and classic APCs such gramming of TAMs with CD40 agonists or PI3Kγ
as dendritic cells (DCs). It is now known that the CNS is not isolated from activated inhibitors73,74, and enhancing TAM phagocytic activity
T cells, which can patrol these compartments in an unrestricted manner, and that with CD47 blockade75, have the potential to be applied
CNS antigens can be presented locally or in the draining cervical lymph nodes. to patients with brain tumours, but each of these
While the immune system in the CNS remains different, it is not incompetent. approaches presents considerable developmental hur-
CSF, cerebrospinal fluid.
dles66. For example, although CD47 antibodies increase
the phagocytosis of tumour cells in mouse xenograft
therapies. Targeting TGFβ with antisense oligonucleo­ models, CD47 ligand affinity in humans differs sub-
tides50 or blocking antibodies51, as well as the use of stantially from that in mice, raising questions about its
TGFβ receptor 1 (TGFβR1) kinase inhibitors52, has failed translational potential76. Thus, while targeting the brain
to show survival benefits. Several ongoing studies are tumour microenvironment represents an attractive ther-
examining the use of IDO inhibitors in primary brain apeutic approach, its implementation may require the
tumours (NCT02502708 (ref.53), NCT02052648 (ref.54), identification of more promising therapeutic targets.
NCT02327078 (ref.55)) (Table 1); however, enthusiasm
for these agents has lessened since the phase III ECHO- Genomic factors
301 trial (NCT02752074 (ref.56)) showed no clinical The most common malignant glioma, glioblastoma,
benefit from adding IDO inhibition to immune check- presents as a genetically heterogeneous disease, despite
point blockade in metastatic melanoma (all secondary a relatively low mutational burden77,78. Clonal selection
sites)57. Several phase I and II clinical trials of arginase of driver mutations occurs as an early event, and it is the
inhibitors for solid tumours are currently underway later acquisition of numerous passenger mutations that
(NCT02903914 (ref.58)), but none is specific for brain results in the intratumoural heterogeneity. Before treat-
tumours. In general, the strategy of inhibiting specific ment, glioblastoma has a neutral evolutionary pattern,
immunosuppressive mediators in patients with brain whereby mutational variants expand at similar rates and
tumours has not shown promise to date, despite its accumulate79. Unfortunately, this situation is lost once
success in animal models, which may be related to the patients commence therapy, when expansion of sub-
penetration of some of these agents into the brain paren- clones is influenced by strong selection pressures and
chyma or the diversity of immunosuppressive mecha- adaptation in response to treatment modalities occurs.
nisms available to the tumour that can compensate for In the setting of low-​g rade glioma, hypermutation
the loss of one. induced by treatment with the alkylating chemothera-
An alternative to targeting specific immunosuppres- peutic agent temozolomide (TMZ) is observed in up to
sive factors is to target the immunosuppressive cells within 60% of tumours at recurrence80. Hypermutations also
the tumour microenvironment that produce such factors. occur in primary glioblastoma at recurrence, but at a
The major cellular component of this microenviron- substantially lower frequency of around 10% of patients
ment is tumour-​associated macrophages (TAMs), which after exposure to TMZ81. The culmination of these

Nature Reviews | Cancer


Reviews

Microglia CCR2 Monocyte T cell clones. These observations may in part explain
antagonists the poor performance of ICIs in the majority of patients
Trabectedin with either newly diagnosed or recurrent glioblastoma.
CCL2 CCR2 Immunoediting is a process whereby the immune sys-
tem eliminates tumour cells carrying immunogenic anti-
CSF1
gens, resulting in the selection of resistant subclones90.
Arginase CSF1R
inhibitors
Arginase TAM
inhibitors
Evidence of this process has been shown in several can-
cer types, including glioma91–95. By comparing newly
diagnosed specimens with those at recurrence from
the same patients, the Glioma Longitudinal Analysis
Arginine Anti-inflammatory TAM Pro-inflammatory TAM (GLASS) Consortium has found evidence in support
depletion of immunoediting in gliomas, suggesting that active
immunosurveillance is ongoing in some adult patients81.
Astrocyte In addition, supporting the possibility of immunoedit-
ing in some patients is a recent study that examined the
• IL-10 Growth • IL-12 whole exome and transcriptome after anti-​programmed
• TGFβ factors • TNF cell death protein 1 (PD1) therapy in recurrent malig-
nant glioma samples. Those tumours defined as respon-
sive were associated with a more diverse pattern of
evolution, possibly due to elimination of neoantigens96.
TGFβ Tumour
inhibitors T cell Killing
cell Several strategies seek to overcome these inherent
mechanisms of resistance. For example, attempts to
identify patients with hypermutated tumours at diag-
nosis are underway, in an effort to enrich for patients
Treg Tryptophan
cell depletion
IDO IDO inhibitors who may respond to immune checkpoint inhibition
(NCT03718767) 97. However, the utility of such an
approach remains unresolved, is perhaps over sim-
Fig. 2 | Immunosuppressive mediators and therapeutic targets in the brain tumour
plistic and may be adversely affected by numerous
microenvironment. Tumour-​associated macrophages (TAMs) play a central role in the
brain tumour microenvironment. TAMs arise from circulating monocytes and, to a lesser
competing factors.
extent, microglia. The recruitment of monocytes and their differentiation into TAMs is Glioblastoma often express a mutated form of the
supported by the chemokine CC-​chemokine ligand 2 (CCL2) and the cytokine colony-​ epidermal growth factor receptor (EGFR), which is
stimulating factor 1 (CSF1). TAMs can be activated towards either an inflammatory or an constitutively active and enhances tumorigenicity by
anti-​inflammatory phenotype. Inflammatory TAMs inhibit tumour growth and support activating the RAS–SHC–GRB2 pathway40. The most
T cell-​mediated tumour killing through the production of inflammatory cytokines such common mutant form of EGFR in glioblastoma is the
as interleukin-12 (IL-12) and tumour necrosis factor (TNF). Anti-​inflammatory TAMs and EGFR class III variant (EGFRvIII), which has a trun-
astrocytes produce IL-10 and transforming growth factor β (TGFβ), both of which inhibit cated extracellular domain owing to an 801 bp in-​frame
T cell effector functions and inflammatory TAM activities45. Anti-​inflammatory TAMs and deletion of the wild-​type receptor98. This deletion results
microglia produce arginase, which inhibits T cells through arginine depletion from the
in the fusion of the two ends of the peptide and the crea-
tumour microenvironment. Gliomas produce indolamine 2,3-dioxygenase (IDO), which
acts to recruit regulatory T (Treg) cells and inhibit effector T cells through tryptophan
tion of an antigenic site that contains a novel glycine res-
depletion. Potential therapeutic strategies to target these immunosuppressive pathways idue not included in the wild-​type peptide99. Therefore,
are shown in purple. CCR2, CC-​chemokine receptor 2; CSF1R , CSF1 receptor. EGFRvIII serves as an ideal tumour-​specific antigen for
glioblastoma. EGFRvIII is expressed in approximately
30% of glioblastomas and is found to be expressed on
events is the rapid emergence of resistance to therapies, 37–86% of tumour cells in a given tissue sample100.
particularly those with a single target82,83. The loss of antigenic targets in response to therapy
In the context of immune-​based therapies, these has severely limited immune-​based therapies that tar-
fundamental observations have a number of major get single antigens. Some of the best examples of this
implications. High tumour mutation burden (TMB) phenomenon are the targeting of EGFRvIII by vaccines
has emerged as a biomarker for immune checkpoint or chimeric antigen receptor (CAR) T cell therapies,
inhibitor (ICI) response, and TMB is believed to cor- in response to which tumours re-​emerge rapidly, hav-
relate with the level of pre-​existing antitumour adaptive ing lost expression of the target, despite documented
immunity84–88. This paradigm applies well to tumours for evidence of CAR T cell infiltration in several patients
which the TMB is greater than 10 mutations/megabase, and preliminary evidence of reduced EGFRvIII tran-
such as melanoma or some colorectal cancers; however, script expression101–103. To overcome this induction of
the median TMB in newly diagnosed glioma is around resistance to single-​target immune-​based therapies,
Lynch syndrome 1.5 mutations/megabase. Fewer than 2% of glioblasto- emerging strategies include those that simultaneously
Also known as hereditary mas from patients in the newly diagnosed setting carry target multiple antigens, such as the recently reported
non-​polyposis colorectal cancer. >10 mutations/megabase, and these usually occur in neoantigen vaccines41,104; those that target ubiquitous
A type of inherited cancer the context of germline mutations of mismatch repair viral antigens, such as those from cytomegalovirus
syndrome associated with a
genetic predisposition to
genes, such as those observed in Lynch syndrome89. Thus, (CMV); and those that aim to induce broad-​b ased
different cancer types, a relatively low TMB offers up few somatic mutations for immune responses, such as innate immune targeting
including glioblastoma. T cell targets and could reduce the array of responding through oncolytic viruses105–109.

www.nature.com/nrc
Reviews

Epigenetic impacts glioblastomas, discovered more recently, that carry SWI/


There is emerging evidence that epigenetic events play a SNF-​related matrix-​associated actin-​dependent regulator
key role in modulating immune responses to tumours, and of chromatin subfamily A-​like protein 1 (SMARCAL1)
many brain tumours carry mutations of epigenetic driv- mutations; and many paediatric tumours, such as atypical
ers110–114. Key examples include isocitrate dehydrogenase teratoid rhabdoid tumour (ATRT) driven by mutations in
(IDH)-mutated gliomas; those carrying histone mutations SMARCB1 (refs110–114). SWI/SNF complexes are impor-
such as in histones H3.1 and H3.3; a subset of primary tant regulators of chromatin structure and transcription

Table 1 | Ongoing clinical trials in brain tumours


Therapy Cancer type Phase nCT identifier
Personalized neoepitope-​based vaccine Recurrent paediatric brain tumours I NCT03068832 (ref.211)
Personalized neoantigen DNA vaccine in combination Newly diagnosed, unmethylated glioblastoma I NCT04015700 (ref.212)
with immune checkpoint blockade therapy
IDO inhibitor (indoximod) in combination with the Progressive primary malignant paediatric brain I NCT02502708 (ref.53)
chemotherapy temozolomide tumours
Anti-​L AG3 +/– anti-​PD1 therapy nivolumab Recurrent glioblastoma I NCT02658981 (ref.168)
Anti-​TIM3 monoclonal (TSR-022) +/– nivolumab Advanced solid tumours I NCT02817633 (ref.169)
Personalized neoantigen-​based vaccine in combination Newly diagnosed, unmethylated glioblastoma I NCT03422094 (ref.213)
with nivolumab
EGFRvIII-​specific CAR T cells in combination with anti-​PD1 Newly diagnosed, unmethylated glioblastoma I NCT03726515 (ref.214)
therapy pembrolizumab
EGFR806-specific CAR T cells in combination with locoregional EGFR-​positive recurrent or refractory I NCT03638167 (ref.215)
immunotherapy (EGFR806 is a monoclonal antibody recognizing paediatric CNS tumours
EGFRvIII and a subset of overexpressed wild-​type EGFRs)
HER2-specific CAR T cells in combination with locoregional HER2-positive recurrent or refractory I NCT03500991 (ref.216)
immunotherapy paediatric CNS tumours
HER2-specific CAR T cells HER2-positive CNS tumours I NCT02442297 (ref.217)
IL-13Rα2-specific CAR T cells Recurrent or refractory malignant glioma I NCT02208362 (ref.218)
CMV pp65 peptide DC vaccine Recurrent medulloblastoma or malignant I NCT03299309 (ref.219)
glioma
CMV pp65 peptide DC vaccine in combination with nivolumab Recurrent brain tumours I NCT02529072 (ref.220)
IDH1 peptide vaccine Recurrent grade II glioma I NCT02193347 (ref.221)
Personalized peptide vaccine in combination with TTFields Glioblastoma I NCT03223103 (ref.222)
Tumour antigen-​loaded DC vaccine Brain tumours I NCT03914768 (ref.223)
IDO inhibitor in combination with temozolomide Primary malignant brain tumours I and II NCT02052648 (ref.54)
IDO inhibitor (epacadostat) in combination with nivolumab Select advanced cancers including brain I and II NCT02327078 (ref.55)
tumours
Arginase inhibitor (INCB001158) +/– immune checkpoint Advanced or metastatic solid tumours I and II NCT02903914 (ref.58)
blockade therapy
Autologous CMV-​specific CTLs in combination with Glioblastoma I and II NCT02661282 (ref.176)
temozolomide
Nivolumab IDH-​mutant gliomas with and without II NCT03718767 (ref.97)
hypermutator phenotype
WT1 peptide vaccine (DSP-7888) in combination with Recurrent or progressive glioblastoma II NCT03149003 (ref.224)
bevacizumab (anti-​VEGFA) following initial therapy
Tumour lysate-​pulsed DC vaccine Brain tumours II NCT01204684 (ref.225)
CMV RNA-​loaded DC vaccine Newly diagnosed WHO Grade IV II NCT03927222 (ref.226)
unmethylated glioma
CMV RNA-​loaded DC vaccine +/– anti-​CD27 therapy Glioblastoma II NCT03688178 (ref.227)
varlilumab (to deplete Treg cells but also enhances activation via
other mechanisms)
Temozolomide in combination with radiation +/– nivolumab Newly diagnosed, unmethylated glioblastoma III NCT02667587 (ref.148)
Nivolumab or temozolomide in combination with radiation Newly diagnosed, unmethylated glioblastoma III NCT02617589 (ref.147)
CAR , chimeric antigen receptor ; CMV, cytomegalovirus; CNS, central nervous system; CTLs, cytotoxic T lymphocytes; DC, dendritic cell; EGFRvIII, epidermal growth
factor receptor variant III; HER2, human epidermal growth factor receptor 2; IDH1, isocitrate dehydrogenase 1; IDO, indolamine 2,3-dioxygenase; IL-13Rα2,
interleukin-13 receptor α2; L AG3, lymphocyte activation gene 3; PD1, programmed cell death protein 1; TIM3, T cell immunoglobulin and mucin domain-​containing
protein 3; Treg, regulatory T; TTFields, tumour-​treating fields; VEGFA , vascular endothelial growth factor A ; WHO, World Health Organization; WT1, Wilms tumour 1.

Nature Reviews | Cancer


Reviews

IDH mutation CD8+ T cell tumour infiltration Immune response


a Repression
CXCL9
Tumour CXCL10
cell
↑2HG T cell

↓CXCL9

Microglia

↓CXCL10 Macrophage

IDH inhibition

b Reprogramming

↓2HG

↑CXCL9

↑CXCL10

Fig. 3 | epigenetic events play a key role in modulating immune responses to brain tumours. a | Isocitrate
dehydrogenase 1 (IDH1) mutations in glioma can cause down-​regulation of leukocyte chemotaxis, resulting in
repression of the tumour-​associated immune system. An IDH mutation results in the generation of the oncometabolite
2-hydroxyglutarate (2HG; via conversion from α-​ketoglutarate), which in turn represses signal transducer and activator
of transcription 1 (STAT1) expression, leading to reduced expression of interferon-​γ (IFNγ)-inducible chemokines,
including CXC chemokine ligand 9 (CXCL9) and CXCL10. As a consequence, IDH-​mutated tumours suppress the
infiltration and accumulation of T cells at tumour sites. b | In mice bearing IDH-​mutated glioma, these effects can be
reversed through pharmacological inhibition with IDH-​C35, a specific inhibitor of mutant IDH. Adapted from ref.228,
Springer Nature Limited.

in several cancers, including brain tumours115. It is now the DNA methyltransferase inhibitor decitabine restores
clear in non-​CNS tumours that mutations of components ULBP1 and ULBP3 expression in IDH-​mutant glioma
within these important protein complexes, such as AT-​ cells122. These data demonstrate a mechanism of immune
rich interactive domain 1A (ARID1A) or polybromo 1 evasion in IDH-​mutated gliomas and suggest that spe-
(PBRM1; also known as BAF180), can drive both resist- cific inhibitors of mutant IDH may improve the effi-
ance and sensitivity to immune-​based therapies through cacy of immunotherapy in patients with IDH-​mutated
alterations in the transcription of downstream targets, gliomas120–122 (Fig. 3).
such as chemokines that attract inflammatory cells116–119. Future studies examining the role of epigenetic mod-
Similar epigenetic events also seem to be at play ulation of immune evasion in malignant gliomas and
in IDH1-mutated glioma. Several groups have shown other brain tumours will be required. It is hypothesized
that IDH1 mutations can cause down-​regulation of that a number of epigenetic drugs, such as demethylating
leukocyte chemotaxis, resulting in repression of the agents, bromodomain inhibitors and histone deacetylase
tumour-​associated immune system120. This is related to inhibitors, may act to reverse the adverse immunologi-
reduced expression of cytotoxic T lymphocyte-​associated cal transcriptional profile seen in epigenetically driven
genes and interferon-​γ (IFNγ)-inducible chemokines, tumours prior to the initiation of immune-​based thera-
including CXC-​chemokine ligand 10 (CXCL10), in pies. These are under investigation preclinically, but as
IDH-​mutated tumours compared with IDH-​wild-type yet they remain unpublished.
tumours121. In syngeneic mouse glioma models, the
expression of mutant IDH1 also suppressed the accu- Impact of standard of care
mulation of T cells in tumour sites via reduced secre- Many patients benefit from multimodal therapy,
tion of CXCL10 (ref.121). In a separate study, IDH mutant including surgery, radiation therapy and chemo-
glioma cells acquired resistance to natural killer (NK) therapy such as TMZ. Although the limitations of
cells through epigenetic silencing of the activating recep- immune-​b ased therapy may be related to tumour-​
tor NKG2D ligands UL16-binding protein 1 (ULBP1) associated factors such as poor immunogenicity and
and ULBP3. In addition, hypomethylation mediated by tumour-​induced immune tolerance, treatment-​induced

www.nature.com/nrc
Reviews

Myelosuppression immunoregulatory effects may also play major roles, showed similarly disappointing results in newly
A condition in which bone both adverse and beneficial. diagnosed patients with O 6-methylguanine-​ DNA-
marrow activity is decreased, Many patients with brain tumours receive adju- methytransferase (MGMT) promoter-​unmethylated
resulting in fewer red blood vant TMZ. Although TMZ provides a survival bene­ glioblastoma. In this study, nivolumab plus radiation
cells, white blood cells and
platelets.
fit in patients with newly diagnosed glioblastoma1, its was compared with the standard combination of radi-
major dose-​limiting toxicity is myelosuppression 123. ation and TMZ. Failure to meet the primary end point
Lymphopenia Furthermore, patients treated with TMZ, especially of OS was announced by Bristol-​Myers Squibb in May of
The condition of having an in dose-​intensified regimens, have an increased inci- 2019, although these results await publication. The
abnormally low level of
dence of opportunistic infections and are immuno­ outcome of CheckMate-548 (NCT02667587 (ref.148)),
lymphocytes in the blood.
suppressed124. Indeed, treatment with the combination which is evaluating nivolumab plus TMZ in patients
of radiation and TMZ is associated with changes in regu­ with MGMT-​methylated glioblastoma, is still pending.
latory and effector mononuclear cells in the peripheral As a consequence, the focus has now shifted towards
blood that tilt the balance towards an immunosuppres- uncovering aetiological contributors to treatment fail-
sive state. This shift could certainly affect the outcome ures, with the goal of removing barriers to successful
of immune-​based therapy following radiation and TMZ immune checkpoint blockade146,149,150.
treatment and should be considered when designing any One question that endures is whether the restric-
combination therapy regimens125,126. However, preclini- tive intracranial environs or simply glioblastoma itself
cal and clinical data have also suggested that a state of presents the greatest challenge to therapeutic efficacy.
lymphopenia upon recovery can induce reactive homeo- A recent clue may be the modest but significant suc-
static proliferation and enhanced antitumour immune cess demonstrated for immune checkpoint strategies
responses. As an example, in a clinical trial of glioblas- targeting other intracranially situated tumours, such as
toma comparing two different radiation and TMZ com- melanoma brain metastases151. Glioblastoma, in turn,
bination regimens, one using standard doses of TMZ does involve its own set of obstacles. The hindrances
at 5 days per month versus a more intensive regimen to immune checkpoint blockade ascribable to glioblas-
of 21 days of TMZ per month, while antigen-​specific toma include a low TMB and extensive intratumoural
immune responses developed in all patients treated with heterogeneity. Even individual cells within glioblas-
either regimen, the more intensive regimen induced toma tumours prove varied in their expression of onco-
an increase in the proportion of immunosuppressive genic transcriptional programmes, reflecting a degree
Treg cells, but also produced humoral and delayed-​type of intratumoural heterogeneity that proffers a unique
hypersensitivity responses of greater magnitude124,127,128.challenge to immunological targeting 9. Additional
Several published studies of vaccine approaches for constraints include the restricted access of drugs
brain tumours now support the hypothesis that lympho- and immune cells to the CNS, as we discussed above.
depletion associated with the dose-​dense 21-of-28-day (However, we should reiterate here that antigen-​specific
TMZ regimen may enhance the immune response, and T cells suffer few constraints to their trafficking26,28,29 and
particularly the humoral immune response to tumour that the BBB is frequently disrupted by CNS tumours
vaccines, which may ultimately translate into improved like glioblastoma30–32.) Nevertheless, what remains less
clinical benefit100,102. Furthermore, data from extracranial
resolved is whether the CNS constraints for drug access
solid tumours129 such as melanoma have explored such or for immune cell access will prove to be more of a
strategies for adoptive immunotherapies. These studies hindrance to effective immune checkpoint blockade.
suggest that well-​timed lymphodepletion may be used For instance, an unresolved question is whether PD1-
to generate positive immunomodulatory effects, in part blocking and CTLA4-blocking antibodies must situ-
owing to the production of an immunostimulatory ate within tumours for their activity, or simply act on
cytokine and chemokine environment130–133. peripheral T cells prior to their CNS entry.
Perhaps the most salient restriction to effective
Current therapeutic approaches immune checkpoint blockade in glioblastoma is ram-
Current approaches, unique observations pant T cell dysfunction, with little baseline T cell acti-
Immune checkpoint inhibitors. Immune checkpoint vation to propagate during therapy37,150,152–156. A viable
blockade targeting the PD1–PD1 ligand 1 (PDL1) axis T  cell compartment is a prerequisite for workable
and/or cytotoxic T lymphocyte-​associated antigen 4 immune checkpoint blockade, which acts primarily to
(CTLA4) has proffered dramatic successes against a unbridle and perpetuate T cell activity. However, T cell
variety of solid tumours134–138 and has garnered US dysfunction has long been identified as a hallmark of
Food and Drug Administration (FDA) approval for a glioblastoma38,155–159, which imposes potent suppression
still increasing number of malignancies139; however, glio- of systemic immunity, despite its intracranial confines.
blastoma remains conspicuously absent from the list of As seen in other solid tumours, resistance to immune
approvals. Although numerous groups have reported checkpoint blockade at the T cell level is marked by the
preclinical successes for more than a decade140–145, upregulation of multiple alternative immune check-
a 2017 phase III trial (CheckMate-143) comparing the points160. These can include T cell immunoglobulin
anti-​PD1 therapy nivolumab with bevacizumab (anti-​ and mucin domain-​containing protein 3 (TIM3), lym-
vascular endothelial growth factor A (VEGFA)) in the phocyte activation gene 3 (LAG3), B and T lymphocyte
treatment of recurrent glioblastoma found no overall attenuator (BTLA), 2B4 (also known as CD244), CD160,
survival (OS) benefit to nivolumab146. More recently, the CD39 and T cell immunoreceptor with immunoglobulin
phase III CheckMate-498 study (NCT02617589 (ref.147)) and ITIM domains (TIGIT)161–163. Mounting expression

Nature Reviews | Cancer


Reviews

of these less canonical immune checkpoints on T cells antigens175. One study examining the administration of
provides further mechanisms for T cell restraint, but CMV-​specific T cells for the treatment of glioblastoma is
it also serves to signal T cell transition into a dysfunc- currently underway (NCT02661282 (ref.176)).
tional, exhausted state163,164. Although exhaustion may be The major advance in adoptive cell therapy in recent
reversible in early stages, it can rapidly progress beyond years has been the development of CAR T cells, which,
recovery165,166. For example, Woroniecka et al.150 recently in the case of CAR T cells targeting CD19, are approved
demonstrated comparatively severe T cell exhaustion for the treatment of B cell leukaemia and lymphoma177.
among TILs isolated from patients and mice with glio- For the treatment of glioblastoma, clinical trial results
blastoma and the corresponding inability of mice with are available for CAR T cells targeting three antigens:
gliomas to respond to PD1 blockade. The increasing EGFRvIII, human epidermal growth factor receptor 2
expression of alternative immune checkpoints may (HER2; also known as ERBB2) and IL-13 receptor α2
indicate a state of terminal exhaustion that cannot be (IL-13Rα2)103,178–180 (Fig. 4). These trials have demon-
reversed by traditional immune checkpoint blockade strated that the use of CAR T cells for brain tumours
alone. However, preclinical work has highlighted a pos- is feasible, safe and potentially efficacious. As with
sible synergy between PD1, TIM3 and LAG3 blockade in other solid tumours, the use of CAR T cells for brain
mice with gliomas167. Clinical trials targeting TIM3 and tumours still faces several substantial obstacles181. One
LAG3, either alone or in combination with anti-​PD1, are major problem is heterogeneous expression of target
underway in glioblastoma (NCT02658981 (ref.168) and antigens in tumour cells. Even in the case of uniformly
NCT02817633 (ref.169)). expressed antigens, selective pressure can result in anti-
Nevertheless, a very recent clinical trial suggests gen loss and tumour recurrence. In the first clinical trial
that the situation may not be as dire as might perhaps of EGFRvIII-​directed CAR T cells for glioblastoma,
be presumed. A simple modification to treatment regi­ a significant decrease in EGFRvIII expression, but not in
mens to include a single neoadjuvant administration wild-​type EGFR, was seen in almost all patients in which
of the anti-​P D1 agent pembrolizumab extended tumour-​infiltrating CAR T cells were observed103. In one
median survival in patients with operable recurrent patient with recurrent multifocal glioblastoma, intra­
glioblastoma to 417 days, compared with 228.5 days cranial administration of IL-13Rα2-targeted CAR T cells
in patients receiving more typical adjuvant dosing resulted in the regression of all intracranial and spinal
of pembrolizumab170. The modification appeared to lesions, but subsequent relapse was due to IL-13Rα2-
capitalize on an improved capacity to generate ante- negative tumours180. This suggests that successful CAR
cedent T cell activation. Likewise, an additional group T cell therapy will require either targeting multiple
demonstrated that neoadjuvant nivolumab precipitated antigens or the development of CAR T cell designs that
pro-​inflammatory changes in the glioblastoma microen- induce significant epitope spreading. Either approach
vironment, although the same clear survival benefits would lead to a broader immune response, which might
were not obtained171. also carry the risk of unintended reactivity against normal
It is clear that glioblastoma poses challenges to tissue. In mouse leukaemia and lymphoma models, CAR
immune checkpoint blockade more paramount than T cells engineered to express CD40L have been shown to
those faced with somewhat more immunogenic tumours, circumvent tumour immune escape via antigen loss by
such as melanoma. While these challenges have likely stimulating CD40–CD40L-​mediated cytotoxicity and the
delayed the advent of truly viable immunotherapeutic induction of robust endogenous immune responses182.
approaches for glioblastoma, a newer focus on mechanis- In an intriguing study using intracranial mouse models
tic considerations for treatment resistance and improving of antigen-​heterogeneous gliomas, Choi et al. examined
the functionality of the T  cell compartment holds tumour responses to EGFRvIII-​directed CAR T cells that
promise for providing important incremental advances were engineered to also express bispecific T cell engag-
to efficacy. ers (BiTEs) targeted to wild-​type EGFR. These BiTE-​
expressing CAR T cells were able to recruit bystander
CARs and adoptive T cells. Adoptive T cell therapy T cells, eliminate heterogeneous tumours and, despite
holds considerable promise for the treatment of the targeting of wild-​type EGFR, displayed no toxicity
brain tumours. Earlier approaches involving non- towards EGFR-​expressing normal tissues183.
specific effector cells, such as NK cells or lymphokine-​ Another substantial issue, associated in particular
activated killer (LAK) cells, have now fallen out of favour, with this type of therapy in brain tumours, is maximiz-
owing to their lack of efficacy172. The administration of ing and maintaining the activity of the administered
autologous TILs has induced regressions in some tumour CAR T cells. CAR T cell administration appears to
types173 but is less feasible in glioblastoma, owing to the induce a compensatory immunosuppressive response
difficulty of isolating and expanding TILs from the CNS. in the brain, characterized by increased expression of
A more feasible approach has been the administration immunosuppressive factors and an influx of Treg cells103.
of autologous CMV-​specific T cells, whereby periph- For this reason, successful therapies will likely require
eral blood mononuclear cells can be isolated from the the development or engineering of CAR T cells resist-
Lymphokine-​activated killer blood and expanded in vitro with synthetic CMV pep- ant to such immunosuppression. Two recent studies of
(LAK) cells tides to generate CMV-​specific T cells, which can then CD19-targeted CAR T cells may be applicable to this
Lymphocytes cultured in the
presence of interleukin-2 (IL-2)
be reinfused into the patient174. This strategy is based problem. Fraietta et al.184 identified factors that were
to stimulate their cytotoxic on the finding that the majority of glioblastomas, but associated with clinical success in patients with chronic
activity. not the surrounding normal brain tissues, express CMV lymphocytic leukaemia (CLL). Clinical responses were

www.nature.com/nrc
Reviews

1 EGFRvIII-directed CAR 2 Release of inflammatory 3 Recruitment of APCs


T cell tumour killing type 1 cytokines, DAMPs and DC engulfment
Tumour and tumour antigens of dying tumours
cell
TH
Immature
DC

TC

Glioblastoma tumour
expressing EGFRvIII

4 DC activation
6 TC and TH newly primed and trafficking
against novel tumour antigens to LN
traffic to and destroy tumour

Mature
5 DCs activate DC
naive T cells
in LN

Glioblastoma
tumour lacking
EGFRvIII Lymph
node

Antigens Antigen derived from EGFRvIII CD4 CD8 CAR

DAMP IFNγ IL-2 MHC-I TCR TNF

Fig. 4 | Chimeric antigen receptor T cell immunotherapy in brain tumours. Chimeric antigen receptor (CAR) T cells
have been used to target tumour-​specific and tumour-​associated antigens in malignant gliomas. There appears to be no
impediment to these cells reaching and killing target antigen-​expressing tumour cells in the central nervous system (CNS).
The challenge remains for these highly specific and potent agents to target antigen-​negative tumour cells directly within
these highly heterogeneous tumours. APC, antigen-​presenting cell; DAMP, damage-​associated molecular pattern;
DC, dendritic cell; EGFRvIII, epidermal growth factor receptor variant III; IFNγ, interferon-​γ; LN, lymph node; MHC-​I, major
histocompatibility complex class I; Tc, cytotoxic T cell; TCR , T cell receptor ; TH, T helper cell; TNF, tumour necrosis factor.
Adapted from ref.229 (Johnson, L. A., Sanchez-​Perez, L., Suryadevara, C. M. & Sampson, J. H. Chimeric antigen receptor
engineered T cells can eliminate brain tumors and initiate long-​term protection against recurrence. Oncoimmunology 3,
e944059 (2014)), with permission of the publisher (Taylor & Francis Ltd, http://www.tandfonline.com).

not associated with any characteristics of the patients in the administered CAR T cells could significantly
or tumours, but rather with the intrinsic properties increase their efficacy. Moreover, pharmacological inhi-
of the administered cells. Markers of response included bition of such enzymes would also have the potential
the expression of memory-​related genes, IL-6–signal to increase the antitumour activity of TAMs, as TET2-
transducer and activator of transcription 3 (STAT3) sig- deficient macrophages have been shown to display
natures, robust ex vivo proliferation and an increased increased inflammatory responses189,190.
frequency of CD27+PD1–CD8+ CAR T cells expressing While CAR T cell strategies remain a popular main-
high levels of the IL-6 receptor (IL-6R). Poor responses stay of engineered T cell therapies for brain tumours,
were associated with the expression of genes involved in alternative strategies exist and are being actively inves-
effector differentiation, glycolysis, exhaustion and apop- tigated. For instance, rather than equipping T cells
tosis. This study strongly suggests that specific features with CAR constructs, cloned T cell receptors (TCRs)
of the CAR T cell product can predict clinical efficacy. possessing a desired antigen specificity can likewise be
This same group also described an extraordinary case transduced into T cells, which are then expanded and
in which complete remission of CLL was attributed to adoptively transferred. Such TCR-​transduced T cells
the massive in vivo expansion of a single CAR T cell remain MHC-​restricted and MHC-​dependent (unlike
clone185. Lentiviral integration of the CAR construct into CARs), but the CAR requirement for surface antigen
the TET2 gene, encoding a methylcytosine dioxygenase, expression is obviated. Such strategies, then, are use-
led to a null mutation of TET2 (ref.185), whose protein ful when targeting intracellular antigens. One recent
product normally regulates the expression of multiple example has been the engineering of a TCR targeting
genes186. TET2-mutated cells have a proliferative advan- a mutated form of the H3 variant H3.3K27M, a com-
tage and display increased inflammatory responses and mon mutation in paediatric diffuse intrinsic pontine
memory T cell differentiation187,188. These findings raise gliomas (DIPGs). T cells transduced with the relevant
the possibility that inhibiting TET2 or similar enzymes TCR demonstrated efficacy when transferred into mice

Nature Reviews | Cancer


Reviews

bearing DIPG xenografts191. Interestingly, H3.3K27M DCVax-L, has advanced to a phase III trial, and prelim-
DIPGs have been found to uniformly express the disialo­ inary results indicate that, on the basis of intent to treat,
ganglioside GD2 at high levels and, in orthotopic DCVax-​L may improve OS203. However, concerns have
H3K27M+-diffuse midline glioma xenograft models, been raised about this report, in that it presents only
GD2-targeted CAR T cells cleared the vast majority interim data, and the primary end point, the number of
of tumours192. progression-free survival events, was not reported.
Methods to improve the antigen-​presenting cell
Vaccines. Therapeutic vaccination for brain tumours (APC) function, cytokine production and migration to
could be a promising potential therapeutic modality lymph nodes of DC vaccines have also been examined.
but is still unproven as yet. Although supportive data Initial work focused on the optimization of conditions
from phase III trials are still lacking, several vaccine for in vitro DC generation in order to maximize DC
strategies have shown good safety and immunogen­ immunogenicity204. The addition of agents such as type I
icity in phase I and II clinical trials, with some vaccines interferon and the synthetic Toll-​like receptor 3 (TLR3)
having demonstrated improved survival relative to ligand polyinosinic:polycytidylic acid (poly(I:C))
historical controls. Peptide vaccines targeting a single improves DC function in vitro205. In phase I and II trials
tumour antigen include those for EGFRvIII, IDH-​ in patients with gliomas, DC vaccines prepared in this
R132H, Wilms tumour 1 (WT1) and survivin. A vac- manner increased post-​vaccine cytokine production and
cine against EGFRvIII in glioblastoma led to substantial were suggested to provide a survival benefit200. An alter-
increases in survival in uncontrolled phase II trials but native strategy has been to improve DC vaccine activity
to no such benefit in a randomized phase III trial193,194. by additional treatments to patients. For example, induc-
In a study of WT1 peptide vaccination, the develop- ing memory T cell activation with tetanus toxoid prior
ment of anti-​WT1 IgG responses was associated with to vaccination with CMV antigen-​loaded DCs increases
increased survival in patients with glioblastoma in a migration of the administered DCs to lymph nodes and
nonrandomized trial195. A substantial problem with appears to improve patient survival206. Other strategies
single-​peptide vaccinations is the potential for tumour tested in humans to increase inflammation at the vaccine
immune escape. In the EGFRvIII vaccine studies, the site have included the administration of granulocyte–
majority of patients who experienced recurrence had macrophage colony-​stimulating factor (GM-​CSF)127,
lost EGFRvIII expression193. For this reason, clinical poly(I:C)200 and other TLR agonists199. In reality, the next
trials have begun to investigate multi-​peptide vaccines major advance in DC vaccines is likely to come from
to target multiple glioma antigens, but none to date has their combination with other immunotherapies. As has
given a clear indication as to efficacy196,197. Two recent been proposed by some207, DC vaccination may be the
trials that have examined the use of personalized pep- best solution to the lack of efficacy seen with immune
tide vaccines in glioma, which are designed according checkpoint blockade in tumours, such as glioblastoma,
to the mutations and gene expression patterns in an with low mutational burdens.
individual patient’s tumour, have provided several novel
observations41,104. First, administration of the corticos- Biomarkers of response
teroid dexamethasone, which is used to reduce cerebral In the context of immune-​based therapies for all can-
oedema during vaccine priming, appeared to inhibit cers, the prediction and monitoring of responses has
systemic antigen-​specific T cell responses. Second, been a major barrier to progress. Tumour biopsy-​based
neoantigen vaccination could induce antigen-​specific techniques for measuring response, such as immunohisto­
TILs, but these cells displayed an exhausted phenotype. chemistry, flow cytometry-​based techniques or sophis-
Third, neoantigen-​specific T cell responses occurred ticated multi-​omic approaches, remain unvalidated
frequently, but primarily among CD4+ T cells, despite and are often limited in their use for brain tumours,
the selection of peptides based on MHC class I bind- owing to the clinical and ethical barriers related to
ing predictions41,104. These findings suggest that corti- longitudinal sample acquisition. Magnetic resonance
costeroids should be avoided during the use of tumour imaging (MRI) has been the main modality used in the
vaccines, that vaccine efficacy may require additional follow-​up and monitoring of treatment response, but
measures to combat T cell exhaustion and that vaccine difficulties arise related to the differentiation between
neoantigens should not be selected solely on the basis pseudoprogression, radiation-​mediated necrosis and
of MHC class I binding predictions. actual tumour progression208,209.
Studies of dendritic cell (DC) vaccines for brain Three recent studies have focused efforts on in-​depth
tumours have examined multiple types of loaded anti- analysis of glioblastoma tissue from patients treated with
gens. Antigen preparations loaded into DCs in clinical ICI therapy in a neoadjuvant setting96,170,171. These data
Pseudoprogression trials have included mixtures of autologous tumour revealed enhanced expression of chemokine transcripts,
A new or enlarging area of
peptides eluted from tumour-​derived cell lines198, autol- higher immune cell infiltration and augmented TCR
contrast agent enhancement
occurring early after the ogous tumour lysates199, peptides from known glioma clonal diversity among TILs, supporting a local immuno­
end of radiotherapy or antigens200 and tumour-​associated viral antigens such modulatory effect of treatment. Additionally, in a
immunotherapy (for example, as CMV127. An initial trial of DCs loaded with six glio- separate study, neoadjuvant PD1 blockade was associ-
within 3–4 months), in the blastoma antigen peptides, termed ICT-107, suggested ated with the upregulation of T cell- and IFNγ-​related
absence of true tumour
growth, that subsides or
there was a survival benefit, but a randomized phase II gene expression in immune cells and the downregu-
stabilizes without a change trial showed no significant improvement in OS201,202. lation of cell-​cycle-related gene expression within the
in therapy. One autologous tumour lysate-​pulsed DC vaccine, tumour96,170,171. Other observations have included focal

www.nature.com/nrc
Reviews

induction of PDL1 in the tumour microenvironment, and rarely have even one mutation that is expressed
enhanced clonal expansion of T cells, decreased PD1 homogeneously. Moreover, the genetic evolution of these
expression on peripheral blood T cells and a decreas- tumours leads to tumour populations that are extremely
ing monocytic population96,170,171. These anti-​PD1 ther- heterogeneous and therefore difficult to target with a
apies appear to lead to different responses in tumours therapy focused on a single antigen. Unfortunately, ther-
with specific molecular alterations. Specifically, non-​ apies targeted at tumour-​associated antigens or multiple
responders are associated with significant enrichment antigens risk unintended consequences to the brain and
of PTEN mutations and immunosuppressive expression systemic tissues alike. The host also provides particular
signatures, whereas responders are associated with an challenges. While the concept of CNS immune privi-
enrichment of MAPK pathway alterations (protein tyro­ lege has eroded over time, there are ways in which the
sine phosphatase, non-​receptor type 11 (PTPN11) and brain treats a foreign body that are simply different from
BRAF), suggesting a genotype–phenotype link41,170,210. those in other organs in the host, and the opportunities
Taken together, such data point to the need for continued we have to provide therapies that access the brain are
development of predictive biomarkers for immune-based limited. In addition, tumours in the brain seem able to
therapies in glioma. promote bone marrow sequestration of the immune cells
that could attack the tumour37. Finally, the standard ther-
Conclusions and perspective apies for this disease, principally radiation and chemo-
Malignant brain tumours, particularly primary gliomas, therapy, are both profoundly immunosuppressive and
remain universally lethal — immunotherapy has done thwart many immunotherapeutic attempts. In the end,
little to improve this. What accounts for these failures? immunotherapy will only be successful if it is specific,
There are likely a complex set of interactions between the can be successfully delivered to the brain and effectively
tumour, its host and the standard therapy used for these deals with cellular heterogeneity.
challenging lesions. These tumours tend to have few
mutations that could be targeted immunotherapeutically Published online xx xx xxxx

1. Stupp, R. et al. Radiotherapy plus concomitant and the brain: a new view. Immunol. Today 13, 507–512 28. Schlager, C. et al. Effector T-​cell trafficking between the
adjuvant temozolomide for glioblastoma. N. Engl. (1992). leptomeninges and the cerebrospinal fluid. Nature
J. Med. 352, 987–996 (2005). 15. Goldmann, J. et al. T cells traffic from brain to cervical 530, 349–353 (2016).
2. Stummer, W. et al. Fluorescence-​guided surgery lymph nodes via the cribroid plate and the nasal 29. Zamvil, S. S. & Steinman, L. The T lymphocyte in
with 5-aminolevulinic acid for resection of malignant mucosa. J. Leukoc. Biol. 80, 797–801 (2006). experimental allergic encephalomyelitis. Annu. Rev.
glioma: a randomised controlled multicentre phase III 16. Widner, H. et al. Scintigraphic method to quantify the Immunol. 8, 579–621 (1990).
trial. Lancet Oncol. 7, 392–401 (2006). passage from brain parenchyma to the deep cervical 30. Long, D. M. Capillary ultrastructure and the blood–
3. Walker, M. D. et al. Randomized comparisons of lymph nodes in rats. Eur. J. Nucl. Med. 13, 456–461 brain barrier in human malignant brain tumors.
radiotherapy and nitrosoureas for the treatment (1987). J. Neurosurg. 32, 127–144 (1970).
of malignant glioma after surgery. N. Engl. J. Med. 17. Aspelund, A. et al. A dural lymphatic vascular system 31. Vajkoczy, P. & Menger, M. D. Vascular microenvironment
303, 1323–1329 (1980). that drains brain interstitial fluid and macromolecules. in gliomas. J. Neurooncol. 50, 99–108 (2000).
4. Hegi, M. E. et al. MGMT gene silencing and benefit J. Exp. Med. 212, 991–999 (2015). 32. Watkins, S. et al. Disruption of astrocyte–vascular
from temozolomide in glioblastoma. N. Engl. J. Med. 18. Louveau, A. et al. CNS lymphatic drainage and coupling and the blood-​brain barrier by invading
352, 997–1003 (2005). neuroinflammation are regulated by meningeal glioma cells. Nat. Commun. 5, 4196 (2014).
5. Imperato, J. P., Paleologos, N. A. & Vick, N. A. lymphatic vasculature. Nat. Neurosci. 21, 1380–1391 33. Li, Q. & Barres, B. A. Microglia and macrophages in
Effects of treatment on long-​term survivors with (2018). brain homeostasis and disease. Nat. Rev. Immunol.
malignant astrocytomas. Ann. Neurol. 28, 818–822 19. Louveau, A. et al. Structural and functional features 18, 225–242 (2018).
(1990). of central nervous system lymphatic vessels. Nature 34. Ginhoux, F. et al. Fate mapping analysis reveals that
6. Gilbert, M. R. et al. A randomized trial of bevacizumab 523, 337–341 (2015). adult microglia derive from primitive macrophages.
for newly diagnosed glioblastoma. N. Engl. J. Med. 20. Eide, P. K., Vatnehol, S. A. S., Emblem, K. E. & Science 330, 841–845 (2010).
370, 699–708 (2014). Ringstad, G. Magnetic resonance imaging provides 35. Ajami, B., Bennett, J. L., Krieger, C., McNagny, K. M.
7. Stupp, R. et al. LTBK-01: prospective, multi-​center evidence of glymphatic drainage from human & Rossi, F. M. Infiltrating monocytes trigger EAE
phase III trial Tumor Treating Fields together brain to cervical lymph nodes. Sci. Rep. 8, 7194 progression, but do not contribute to the resident
temozolomide compared temozolomide alone patients (2018). microglia pool. Nat. Neurosci. 14, 1142–1149 (2011).
newly diagnosed glioblastoma. Neuro Oncol. 18 21. Iliff, J. J. et al. A paravascular pathway facilitates 36. Bennett, F. C. et al. A combination of ontogeny and
(Suppl. 6), i1 (2016). CSF flow through the brain parenchyma and the CNS environment establishes microglial identity.
8. Stupp, R. et al. Effect of tumor-​treating fields plus clearance of interstitial solutes, including amyloid Neuron 98, 1170–1183 e1178 (2018).
maintenance temozolomide vs maintenance beta. Sci. Transl. Med. 4, 147ra111 (2012). 37. Chongsathidkiet, P. et al. Sequestration of T cells in
temozolomide alone on survival in patients with This research was the first to document the bone marrow in the setting of glioblastoma and other
glioblastoma: a randomized clinical trial. JAMA 318, existence of the glymphatic pathway. intracranial tumors. Nat. Med. 24, 1459–1468
2306–2316 (2017). 22. Kipnis, J. Multifaceted interactions between adaptive (2018).
9. Patel, A. P. et al. Single-​cell RNA-​seq highlights immunity and the central nervous system. Science This research uncovers the novel phenomenon
intratumoral heterogeneity in primary glioblastoma. 353, 766–771 (2016). of T cell sequestration within the bone marrow.
Science 344, 1396–1401 (2014). 23. Bechmann, I., Galea, I. & Perry, V. H. What is the The phenomenon is specific to tumours of the
This study provides one of the most thorough blood–brain barrier (not)? Trends Immunol. 28, 5–11 intracranial compartment and suggests a novel
documentations to date of glioblastoma (2007). mechanism for CNS-​imposed limitations to T cell
heterogeneity at the single-​cell level. 24. Huber, J. D., Egleton, R. D. & Davis, T. P. Molecular access that tumours can usurp.
10. Vogelstein, B. et al. Cancer genome landscapes. physiology and pathophysiology of tight junctions in 38. Fecci, P. E., Heimberger, A. B. & Sampson, J. H.
Science 339, 1546–1558 (2013). the blood-​brain barrier. Trends Neurosci. 24, Immunotherapy for primary brain tumors: no longer a
11. Engelhardt, B., Vajkoczy, P. & Weller, R. O. 719–725 (2001). matter of privilege. Clin. Cancer Res. 20, 5620–5629
The movers and shapers in immune privilege 25. Tietz, S. & Engelhardt, B. Brain barriers: crosstalk (2014).
of the CNS. Nat. Immunol. 18, 123–131 (2017). between complex tight junctions and adherens 39. Gajewski, T. F., Schreiber, H. & Fu, Y. X. Innate and
12. Medawar, P. B. Immunity to homologous grafted skin; junctions. J. Cell Biol. 209, 493–506 (2015). adaptive immune cells in the tumor microenvironment.
the fate of skin homografts transplanted to the brain, 26. Owens, T., Bechmann, I. & Engelhardt, B. Perivascular Nat. Immunol. 14, 1014–1022 (2013).
to subcutaneous tissue, and to the anterior chamber spaces and the two steps to neuroinflammation. 40. Gajewski, T. F. et al. Cancer immunotherapy targets
of the eye. Br. J. Exp. Pathol. 29, 58–69 (1948). J. Neuropathol. Exp. Neurol. 67, 1113–1121 based on understanding the T cell-​inflamed versus
This landmark paper is often credited as the first (2008). non-​T cell-​inflamed tumor microenvironment.
to suggest the notion of CNS immune privilege. Along with Aspelund et al. (2015), this is the first Adv. Exp. Med. Biol. 1036, 19–31 (2017).
13. Bradbury, M. W. & Westrop, R. J. Factors influencing study to characterize CNS lymphatic vessels within 41. Keskin, D. B. et al. Neoantigen vaccine generates
exit of substances from cerebrospinal fluid into deep the meninges. intratumoral T cell responses in phase Ib glioblastoma
cervical lymph of the rabbit. J. Physiol. 339, 519–534 27. Andersson, P. B., Perry, V. H. & Gordon, S. The acute trial. Nature 565, 234–239 (2019).
(1983). inflammatory response to lipopolysaccharide in CNS This study highlights some issues associated
14. Cserr, H. F. & Knopf, P. M. Cervical lymphatics, the parenchyma differs from that in other body tissues. with using the neoantigen vaccine strategy
blood–brain barrier and the immunoreactivity of Neuroscience 48, 169–186 (1992). for glioblastoma.

Nature Reviews | Cancer


Reviews

42. Quail, D. F. & Joyce, J. A. The microenvironmental 68. Pyonteck, S. M. et al. CSF-1R inhibition alters immunosurveillance to tumor escape. Nat. Immunol.
landscape of brain tumors. Cancer Cell 31, 326–341 macrophage polarization and blocks glioma 3, 991–998 (2002).
(2017). progression. Nat. Med. 19, 1264–1272 (2013). 96. Zhao, J. et al. Immune and genomic correlates of
This excellent review article includes an in-​depth 69. US National Library of Medicine. ClinicalTrials.gov, response to anti-​PD-1 immunotherapy in glioblastoma.
discussion of the brain tumour microenvironment, https://ClinicalTrials.gov/show/NCT01349036 (2011). Nat. Med. 25, 462–469 (2019).
relevant cell types, potential tumour-​promoting 70. Butowski, N. et al. Orally administered colony 97. US National Library of Medicine. ClinicalTrials.gov,
mechanisms and possible therapeutics. stimulating factor 1 receptor inhibitor PLX3397 in https://ClinicalTrials.gov/show/NCT03718767 (2018).
43. Perng, P. & Lim, M. Immunosuppressive mechanisms recurrent glioblastoma: an Ivy Foundation Early Phase 98. Bigner, S. H. et al. Characterization of the epidermal
of malignant gliomas: parallels at non-​CNS sites. Clinical Trials Consortium phase II study. Neuro Oncol. growth factor receptor in human glioma cell lines and
Front. Oncol. 5, 153 (2015). 18, 557–564 (2016). xenografts. Cancer Res. 50, 8017–8022 (1990).
44. Vitkovic, L., Maeda, S. & Sternberg, E. Anti-​ 71. Germano, G. et al. Role of macrophage targeting in 99. Humphrey, P. A. et al. Anti-​synthetic peptide antibody
inflammatory cytokines: expression and action in the antitumor activity of trabectedin. Cancer Cell 23, reacting at the fusion junction of deletion-​mutant
the brain. Neuroimmunomodulation 9, 295–312 249–262 (2013). epidermal growth factor receptors in human
(2001). 72. Vakilian, A., Khorramdelazad, H., Heidari, P., glioblastoma. Proc. Natl Acad. Sci. USA 87,
45. Gong, D. et al. TGFbeta signaling plays a critical role Sheikh Rezaei, Z. & Hassanshahi, G. CCL2/CCR2 4207–4211 (1990).
in promoting alternative macrophage activation. signaling pathway in glioblastoma multiforme. 100. Wikstrand, C. J., McLendon, R. E., Friedman, A. H.
BMC Immunol. 13, 31 (2012). Neurochem. Int. 103, 1–7 (2017). & Bigner, D. D. Cell surface localization and density of
46. Wainwright, D. A. et al. IDO expression in brain 73. Vonderheide, R. H. & Glennie, M. J. Agonistic CD40 the tumor-​associated variant of the epidermal growth
tumors increases the recruitment of regulatory T cells antibodies and cancer therapy. Clin. Cancer Res. 19, factor receptor, EGFRvIII. Cancer Res. 57, 4130–4140
and negatively impacts survival. Clin. Cancer Res. 18, 1035–1043 (2013). (1997).
6110–6121 (2012). 74. Kaneda, M. M. et al. PI3Kgamma is a molecular 101. Padfield, E., Ellis, H. P. & Kurian, K. M. Current
47. Uyttenhove, C. et al. Evidence for a tumoral immune switch that controls immune suppression. Nature Therapeutic Advances Targeting EGFR and EGFRvIII
resistance mechanism based on tryptophan degradation 539, 437–442 (2016). in Glioblastoma. Front. Oncol. 5, 5 (2015).
by indoleamine 2,3-dioxygenase. Nat. Med. 9, 75. Weiskopf, K. Cancer immunotherapy targeting the 102. Migliorini, D. et al. CAR T-​Cell Therapies in glioblastoma:
1269–1274 (2003). CD47/SIRPalpha axis. Eur. J. Cancer 76, 100–109 a first look. Clin. Cancer Res. 24, 535–540 (2018).
48. Zhang, I. et al. Characterization of arginase expression (2017). 103. O'Rourke, D. M. et al. A single dose of peripherally
in glioma-​associated microglia and macrophages. 76. Kwong, L. S., Brown, M. H., Barclay, A. N. infused EGFRvIII-​directed CAR T cells mediates
PLOS ONE 11, e0165118 (2016). & Hatherley, D. Signal-​regulatory protein alpha from the antigen loss and induces adaptive resistance in
49. Hawkins, R. A., O'Kane, R. L., Simpson, I. A. NOD mouse binds human CD47 with an exceptionally patients with recurrent glioblastoma. Sci. Transl. Med.
& Vina, J. R. Structure of the blood–brain barrier and high affinity—implications for engraftment of human 9, eaaa0984 (2017).
its role in the transport of amino acids. J. Nutr. 136, cells. Immunology 143, 61–67 (2014). 104. Hilf, N. et al. Actively personalized vaccination trial for
218S–226S (2006). 77. Hodges, T. R. et al. Mutational burden, immune newly diagnosed glioblastoma. Nature 565, 240–245
50. Bogdahn, U. et al. Targeted therapy for high-​grade checkpoint expression, and mismatch repair in glioma: (2019).
glioma with the TGF-​beta2 inhibitor trabedersen: implications for immune checkpoint immunotherapy. 105. Cloughesy, T. F. et al. Phase 1 trial of vocimagene
results of a randomized and controlled phase IIb study. Neuro Oncol. 19, 1047–1057 (2017). amiretrorepvec and 5-fluorocytosine for recurrent
Neuro Oncol. 13, 132–142 (2011). 78. Bouffet, E. et al. Immune checkpoint inhibition for high-​grade glioma. Sci. Transl. Med. 8, 341ra375
51. den Hollander, M. W. et al. TGF-​beta antibody uptake hypermutant glioblastoma multiforme resulting (2016).
in recurrent high-​grade glioma imaged with 89Zr-​ from germline biallelic mismatch repair deficiency. 106. Desjardins, A. et al. Recurrent glioblastoma treated
fresolimumab PET. J. Nucl. Med. 56, 1310–1314 J. Clin. Oncol. 34, 2206–2211 (2016). with recombinant poliovirus. N. Engl. J. Med. 379,
(2015). 79. Muscat, A. M. et al. The evolutionary pattern of 150–161 (2018).
52. Brandes, A. A. et al. A phase II randomized study mutations in glioblastoma reveals therapy-​mediated 107. Fountzilas, C., Patel, S. & Mahalingam, D. Review:
of galunisertib monotherapy or galunisertib plus selection. Oncotarget 9, 7844–7858 (2018). oncolytic virotherapy, updates and future directions.
lomustine compared with lomustine monotherapy in 80. Johnson, B. E. et al. Mutational analysis reveals the Oncotarget 8, 102617–102639 (2017).
patients with recurrent glioblastoma. Neuro Oncol. origin and therapy-​driven evolution of recurrent 108. Lang, F. F. et al. Phase I study of DNX-2401
18, 1146–1156 (2016). glioma. Science 343, 189–193 (2014). (Delta-24-RGD) oncolytic adenovirus: replication and
53. US National Library of Medicine. ClinicalTrials.gov, 81. Barthel, F. et al. Longitudinal molecular trajectories immunotherapeutic effects in recurrent malignant
https://ClinicalTrials.gov/show/NCT02502708 (2015). of diffuse glioma in adults. Nature (in the press). glioma. J. Clin. Oncol. 36, 1419–1427 (2018).
54. US National Library of Medicine. ClinicalTrials.gov, 82. Kim, H. et al. Whole-​genome and multisector exome 109. Markert, J. M. et al. A phase 1 trial of oncolytic
https://ClinicalTrials.gov/show/NCT02052648 (2014). sequencing of primary and post-​treatment glioblastoma HSV-1, G207, given in combination with radiation for
55. US National Library of Medicine. ClinicalTrials.gov, reveals patterns of tumor evolution. Genome Res. 25, recurrent GBM demonstrates safety and radiographic
https://ClinicalTrials.gov/show/NCT02327078 (2014). 316–327 (2015). responses. Mol. Ther. 22, 1048–1055 (2014).
56. US National Library of Medicine. ClinicalTrials.gov, 83. Wang, J. et al. Clonal evolution of glioblastoma under 110. Cai, J. et al. Loss of ATRX, associated with DNA
https://ClinicalTrials.gov/show/NCT02752074 (2016). therapy. Nat. Genet. 48, 768–776 (2016). methylation pattern of chromosome end, impacted
57. Long, G. V. et al. Epacadostat (E) plus pembrolizumab 84. Schumacher, T. N. & Schreiber, R. D. Neoantigens biological behaviors of astrocytic tumors. Oncotarget
(P) versus pembrolizumab alone in patients (pts) with in cancer immunotherapy. Science 348, 69–74 6, 18105–18115 (2015).
unresectable or metastatic melanoma: results of the (2015). 111. Gusyatiner, O. & Hegi, M. E. Glioma epigenetics:
phase 3 ECHO-301/KEYNOTE-252 study. J. Clin. Oncol. 85. Van Allen, E. M. et al. Genomic correlates of response from subclassification to novel treatment options.
36, 108–108 (2018). to CTLA-4 blockade in metastatic melanoma. Science Semin. Cancer Biol. 51, 50–58 (2018).
58. US National Library of Medicine. ClinicalTrials.gov, 350, 207–211 (2015). 112. Hassan, A., Mosley, J., Singh, S. & Zinn, P. O.
https://ClinicalTrials.gov/show/NCT02903914 86. Goodman, A. M. et al. Tumor mutational burden as an A comprehensive review of genomics and noncoding
(2016). independent predictor of response to immunotherapy RNA in gliomas. Top. Magn. Reson. Imaging 26, 3–14
59. Graeber, M. B., Scheithauer, B. W. & Kreutzberg, G. W. in diverse cancers. Mol. Cancer Ther. 16, 2598–2608 (2017).
Microglia in brain tumors. Glia 40, 252–259 (2002). (2017). 113. Johann, P. D. et al. Atypical teratoid/rhabdoid tumors are
60. Chen, Z. et al. Cellular and molecular identity of 87. Hellmann, M. D. et al. Nivolumab plus ipilimumab in comprised of three epigenetic subgroups with distinct
tumor-​associated macrophages in glioblastoma. lung cancer with a high tumor mutational burden. enhancer landscapes. Cancer Cell 29, 379–393 (2016).
Cancer Res. 77, 2266–2278 (2017). N. Engl. J. Med. 378, 2093–2104 (2018). 114. Mao, C. X. et al. The molecular classification of
61. Bowman, R. L. et al. Macrophage ontogeny underlies 88. Hellmann, M. D. et al. Genomic features of response astrocytic tumors. Oncotarget 8, 96340–96350
differences in tumor-​specific education in brain to combination immunotherapy in patients with (2017).
malignancies. Cell Rep. 17, 2445–2459 (2016). advanced non-​small-cell lung cancer. Cancer Cell 33, 115. Loo Yau, H., Ettayebi, I. & De Carvalho, D. D.
62. Zhou, W. et al. Periostin secreted by glioblastoma stem 843–852.e844 (2018). The cancer epigenome: exploiting its vulnerabilities
cells recruits M2 tumour-​associated macrophages and 89. Therkildsen, C. et al. Glioblastomas, astrocytomas and for immunotherapy. Trends Cell Biol. 29, 31–43
promotes malignant growth. Nat. Cell Biol. 17, oligodendrogliomas linked to Lynch syndrome. Eur. J. (2019).
170–182 (2015). Neurol. 22, 717–724 (2015). 116. Pan, D. et al. A major chromatin regulator determines
63. Hambardzumyan, D., Gutmann, D. H. & Kettenmann, H. 90. Dunn, G. P., Old, L. J. & Schreiber, R. D. The three Es resistance of tumor cells to T cell-​mediated killing.
The role of microglia and macrophages in glioma of cancer immunoediting. Annu. Rev. Immunol. 22, Science 359, 770–775 (2018).
maintenance and progression. Nat. Neurosci. 19, 329–360 (2004). 117. Miao, D. et al. Genomic correlates of response to
20–27 (2016). 91. Marty Pyke, R. et al. Evolutionary pressure against immune checkpoint therapies in clear cell renal cell
64. Komohara, Y., Ohnishi, K., Kuratsu, J. & Takeya, M. MHC class II binding cancer mutations. Cell 175, carcinoma. Science 359, 801–806 (2018).
Possible involvement of the M2 anti-​inflammatory 416–428.e413 (2018). 118. Shu, X. S. et al. The epigenetic modifier PBRM1
macrophage phenotype in growth of human gliomas. 92. McGranahan, N. et al. Allele-​Specific HLA loss and restricts the basal activity of the innate immune system
J. Pathol. 216, 15–24 (2008). immune escape in lung cancer evolution. Cell 171, by repressing retinoic acid-​inducible gene-​I-like receptor
65. Murray, P. J. et al. Macrophage activation and 1259–1271 e1211 (2017). signalling and is a potential prognostic biomarker for
polarization: nomenclature and experimental 93. Wang, Q. et al. Tumor evolution of glioma-​intrinsic colon cancer. J. Pathol. 244, 36–48 (2018).
guidelines. Immunity 41, 14–20 (2014). gene expression subtypes associates with 119. Shen, J. et al. ARID1A deficiency promotes mutability
66. Majety, M., Runza, V., Lehmann, C., Hoves, S. immunological changes in the microenvironment. and potentiates therapeutic antitumor immunity
& Ries, C. H. A drug development perspective on Cancer Cell 32, 42–56 e46 (2017). unleashed by immune checkpoint blockade. Nat. Med.
targeting tumor-​associated myeloid cells. FEBS J. 94. Rooney, M. S., Shukla, S. A., Wu, C. J., Getz, G. & 24, 556–562 (2018).
285, 763–776 (2018). Hacohen, N. Molecular and genetic properties of 120. Amankulor, N. M. et al. Mutant IDH1 regulates
67. Hume, D. A. & MacDonald, K. P. Therapeutic tumors associated with local immune cytolytic activity. the tumor-​associated immune system in gliomas.
applications of macrophage colony-​stimulating Cell 160, 48–61 (2015). Genes Dev. 31, 774–786 (2017).
factor-1 (CSF-1) and antagonists of CSF-1 receptor 95. Dunn, G. P., Bruce, A. T., Ikeda, H., Old, L. J. 121. Kohanbash, G. et al. Isocitrate dehydrogenase
(CSF-1R) signaling. Blood 119, 1810–1820 (2012). & Schreiber, R. D. Cancer immunoediting: from mutations suppress STAT1 and CD8+ T cell

www.nature.com/nrc
Reviews

accumulation in gliomas. J. Clin. Invest. 127, intracranial gliomas. Int. J. Radiat. Oncol. Biol. Phys. 170. Cloughesy, T. F. et al. Neoadjuvant anti-​PD-1
1425–1437 (2017). 86, 343–349 (2013). immunotherapy promotes a survival benefit with
122. Zhang, X. et al. IDH mutant gliomas escape natural 146. Filley, A. C., Henriquez, M. & Dey, M. Recurrent intratumoral and systemic immune responses in
killer cell immune surveillance by downregulation of glioma clinical trial, CheckMate-143: the game is not recurrent glioblastoma. Nat. Med. 25, 477–486
NKG2D ligand expression. Neuro Oncol. 18, over yet. Oncotarget 8, 91779–91794 (2017). (2019).
1402–1412 (2016). 147. US National Library of Medicine. ClinicalTrials.gov, This report describes the first trial to show the
123. Su, Y. B. et al. Selective CD4+ lymphopenia in https://ClinicalTrials.gov/show/NCT02617589 (2015). efficacy of immune checkpoint blockade in patients
melanoma patients treated with temozolomide: a 148. US National Library of Medicine. ClinicalTrials.gov, with glioblastoma, which was accomplished via
toxicity with therapeutic implications. J. Clin. Oncol. https://ClinicalTrials.gov/show/NCT02667587 (2016). simple neoadjuvant administration prior to
22, 610–616 (2004). 149. Wu, A. et al. Combination anti-​CXCR4 and anti-​PD-1 resection of recurrent tumours.
124. Sampson, J. H. et al. Greater chemotherapy-​induced immunotherapy provides survival benefit in 171. Schalper, K. A. et al. Neoadjuvant nivolumab modifies
lymphopenia enhances tumor-​specific immune glioblastoma through immune cell modulation of the tumor immune microenvironment in resectable
responses that eliminate EGFRvIII-​expressing tumor tumor microenvironment. J. Neurooncol. 143, glioblastoma. Nat. Med. 25, 470–476 (2019).
cells in patients with glioblastoma. Neuro Oncol. 13, 241–249 (2019). 172. Law, T. M. et al. Phase III randomized trial of
324–333 (2011). 150. Woroniecka, K. et al. T-​Cell exhaustion signatures vary interleukin-2 with or without lymphokine-​activated
125. Fadul, C. E. et al. Immune modulation effects of with tumor type and are severe in glioblastoma. killer cells in the treatment of patients with advanced
concomitant temozolomide and radiation therapy on Clin. Cancer Res. 24, 4175–4186 (2018). renal cell carcinoma. Cancer 76, 824–832 (1995).
peripheral blood mononuclear cells in patients with This study is the first to reveal and characterize the 173. Rosenberg, S. A. & Dudley, M. E. Cancer regression in
glioblastoma multiforme. Neuro Oncol. 13, 393–400 severity of bona fide T cell exhaustion in patients patients with metastatic melanoma after the transfer
(2011). with glioblastoma. of autologous antitumor lymphocytes. Proc. Natl
126. Grossman, S. A. et al. Survival in patients with 151. Tawbi, H. A. et al. Combined nivolumab and ipilimumab Acad. Sci. USA 101, 14639–14645 (2004).
severe lymphopenia following treatment with in melanoma metastatic to the brain. N. Engl. J. Med. 174. Schuessler, A. et al. Autologous T-​cell therapy for
radiation and chemotherapy for newly diagnosed solid 379, 722–730 (2018). cytomegalovirus as a consolidative treatment
tumors. J. Natl Compr. Canc. Netw. 13, 1225–1231 152. Brooks, W. H., Caldwell, H. D. & Mortara, R. H. for recurrent glioblastoma. Cancer Res. 74,
(2015). Immune responses in patients with gliomas. 3466–3476 (2014).
127. Batich, K. A. et al. Long-​term survival in glioblastoma Surg. Neurol. 2, 419–423 (1974). 175. Cobbs, C. S. et al. Human cytomegalovirus infection
with cytomegalovirus pp65-targeted vaccination. 153. Brooks, W. H., Roszman, T. L. & Rogers, A. S. and expression in human malignant glioma. Cancer
Clin. Cancer Res. 23, 1898–1909 (2017). Impairment of rosette-​forming T lymphocytes in Res. 62, 3347–3350 (2002).
128. Asavaroengchai, W., Kotera, Y. & Mule, J. J. Tumor patients with primary intracranial tumors. Cancer 37, 176. US National Library of Medicine. ClinicalTrials.gov,
lysate-​pulsed dendritic cells can elicit an effective 1869–1873 (1976). https://ClinicalTrials.gov/show/NCT02661282 (2016).
antitumor immune response during early lymphoid 154. Fecci, P. E. et al. Increased regulatory T-​cell fraction 177. Maus, M. V., Grupp, S. A., Porter, D. L. & June, C. H.
recovery. Proc. Natl Acad. Sci. USA 99, 931–936 amidst a diminished CD4 compartment explains Antibody-​modified T cells: CARs take the front seat for
(2002). cellular immune defects in patients with malignant hematologic malignancies. Blood 123, 2625–2635
129. Nistico, P. et al. Chemotherapy enhances vaccine-​ glioma. Cancer Res. 66, 3294–3302 (2006). (2014).
induced antitumor immunity in melanoma patients. This publication is the first to demonstrate the 178. Ahmed, N. et al. HER2-specific chimeric antigen
Int. J. Cancer 124, 130–139 (2009). systemic immune impact of Treg cells in patients receptor-​modified virus-​specific T cells for progressive
130. Schaaf, M. B., Garg, A. D. & Agostinis, P. Defining the with glioblastoma. glioblastoma: a phase 1 dose-​escalation trial. JAMA
role of the tumor vasculature in antitumor immunity 155. Dunn, G. P., Fecci, P. E. & Curry, W. T. Cancer Oncol. 3, 1094–1101 (2017).
and immunotherapy. Cell Death Dis. 9, 115 (2018). immunoediting in malignant glioma. Neurosurgery 179. Brown, C. E. et al. Bioactivity and safety of IL13R
131. Yang, J., Yan, J. & Liu, B. Targeting VEGF/VEGFR to 71, 201–222; discussion 222–223 (2012). alpha2-redirected chimeric antigen receptor CD8+
Modulate Antitumor Immunity. Front. Immunol. 9, 156. Woroniecka, K. I., Rhodin, K. E., Chongsathidkiet, P., T cells in patients with recurrent glioblastoma.
978 (2018). Keith, K. A. & Fecci, P. E. T-​cell dysfunction in Clin. Cancer Res. 21, 4062–4072 (2015).
132. Manzoni, M. et al. Immunological effects of glioblastoma: applying a new framework. Clin. Cancer 180. Brown, C. E. et al. Regression of glioblastoma after
bevacizumab-​based treatment in metastatic colorectal Res. 24, 3792–3802 (2018). chimeric antigen receptor T-​cell therapy. N. Engl.
cancer. Oncology 79, 187–196 (2010). 157. Dix, A. R., Brooks, W. H., Roszman, T. L. J. Med. 375, 2561–2569 (2016).
133. Martino, E. C. et al. Immune-​modulating effects of & Morford, L. A. Immune defects observed in patients 181. Long, K. B. et al. CAR T cell therapy of non-​
bevacizumab in metastatic non-​small-cell lung cancer with primary malignant brain tumors. J. Neuroimmunol. hematopoietic malignancies: detours on the road
patients. Cell Death Discov. 2, 16025 (2016). 100, 216–232 (1999). to clinical success. Front. Immunol. 9, 2740 (2018).
134. Larkin, J., Hodi, F. S. & Wolchok, J. D. Combined 158. Brooks, W. H., Netsky, M. G., Normansell, D. E. & 182. Kuhn, N. F. et al. CD40 ligand-​modified chimeric
Nivolumab and Ipilimumab or monotherapy in Horwitz, D. A. Depressed cell-​mediated immunity in antigen receptor T cells enhance antitumor function
untreated melanoma. N. Engl. J. Med. 373, patients with primary intracranial tumors. by eliciting an endogenous antitumor response.
1270–1271 (2015). Characterization of a humoral immunosuppressive Cancer Cell 35, 473–488 e476 (2019).
135. Robert, C. et al. Pembrolizumab versus Ipilimumab factor. J. Exp. Med. 136, 1631–1647 (1972). This article describes a preclinical study in
in advanced melanoma. N. Engl. J. Med. 372, 159. Roszman, T. L. & Brooks, W. H. Immunobiology of which the expression of CD40L on CAR T cells led
2521–2532 (2015). primary intracranial tumours. III. Demonstration of a to the killing of antigen-​negative tumour cells and
136. Motzer, R. J. et al. Nivolumab versus everolimus in qualitative lymphocyte abnormality in patients with the elimination of tumour immune escape via
advanced renal-​cell carcinoma. N. Engl. J. Med. 373, primary brain tumours. Clin. Exp. Immunol. 39, antigen loss.
1803–1813 (2015). 395–402 (1980). 183. Choi, B. D. et al. CAR-​T cells secreting BiTEs circumvent
137. Garon, E. B. et al. Pembrolizumab for the treatment 160. Koyama, S. et al. Adaptive resistance to therapeutic antigen escape without detectable toxicity. Nat.
of non-​small-cell lung cancer. N. Engl. J. Med. 372, PD-1 blockade is associated with upregulation of Biotechnol. 37, 1049–1058 (2019).
2018–2028 (2015). alternative immune checkpoints. Nat. Commun. 7, 184. Fraietta, J. A. et al. Determinants of response and
138. Ferris, R. L. et al. Nivolumab for recurrent squamous-​ 10501 (2016). resistance to CD19 chimeric antigen receptor (CAR)
cell carcinoma of the head and neck. N. Engl. J. Med. 161. Chauvin, J. M. et al. TIGIT and PD-1 impair tumor T cell therapy of chronic lymphocytic leukemia.
375, 1856–1867 (2016). antigen-​specific CD8+ T cells in melanoma patients. Nat. Med. 24, 563–571 (2018).
139. Immunotherapy Timeline of Progress, https://www. J. Clin. Invest. 125, 2046–2058 (2015). This study identifies predictors of clinical response
cancerresearch.org/immunotherapy/timeline-​ 162. Blackburn, S. D. et al. Coregulation of CD8+ T cell to CAR T cell therapy as being exclusively intrinsic
of-progress (2019). exhaustion by multiple inhibitory receptors during properties of the administered cells.
140. Agarwalla, P., Barnard, Z., Fecci, P., Dranoff, G. chronic viral infection. Nat. Immunol. 10, 29–37 185. Fraietta, J. A. et al. Disruption of TET2 promotes the
& Curry, W. T. Jr Sequential immunotherapy by (2009). therapeutic efficacy of CD19-targeted T cells. Nature
vaccination with GM-​CSF-expressing glioma cells and 163. Woo, S. R. et al. Immune inhibitory molecules LAG-3 558, 307–312 (2018).
CTLA-4 blockade effectively treats established murine and PD-1 synergistically regulate T-​cell function to This case report describes how fortuitous deletion of
intracranial tumors. J. Immunother. 35, 385–389 promote tumoral immune escape. Cancer Res. 72, the TET2 gene by CARs led to complete remission
(2012). 917–927 (2012). of CLL.
141. Fecci, P. E. et al. Systemic CTLA-4 blockade ameliorates 164. Kurtulus, S. et al. TIGIT predominantly regulates the 186. Ichiyama, K. et al. The methylcytosine dioxygenase
glioma-​induced changes to the CD4+ T cell compartment immune response via regulatory T cells. J. Clin. Invest. Tet2 promotes DNA demethylation and activation
without affecting regulatory T-​cell function. Clin. Cancer 125, 4053–4062 (2015). of cytokine gene expression in T cells. Immunity 42,
Res. 13, 2158–2167 (2007). 165. Angelosanto, J. M., Blackburn, S. D., Crawford, A. 613–626 (2015).
This study is the first to preclinically test immune & Wherry, E. J. Progressive loss of memory T cell 187. Shlush, L. I. Age-​related clonal hematopoiesis. Blood
checkpoint blockade in a brain tumour model. potential and commitment to exhaustion during 131, 496–504 (2018).
142. Antonios, J. P. et al. PD-1 blockade enhances the chronic viral infection. J. Virol. 86, 8161–8170 188. Carty, S. A. et al. The loss of TET2 promotes CD8+
vaccination-​induced immune response in glioma. (2012). T cell memory differentiation. J. Immunol. 200,
JCI Insight 1, e87059 (2016). 166. Wherry, E. J. & Kurachi, M. Molecular and cellular 82–91 (2018).
143. Reardon, D. A. et al. Glioblastoma eradication following insights into T cell exhaustion. Nat. Rev. Immunol. 189. Cull, A. H., Snetsinger, B., Buckstein, R., Wells, R. A. &
immune checkpoint blockade in an orthotopic, 15, 486–499 (2015). Rauh, M. J. Tet2 restrains inflammatory gene expression
immunocompetent model. Cancer Immunol. Res. 4, 167. Kim, J. E. et al. Combination therapy with anti-​PD-1, in macrophages. Exp. Hematol. 55, 56–70 e13 (2017).
124–135 (2016). anti-​TIM-3, and focal radiation results in regression 190. Zhang, Q. et al. Tet2 is required to resolve
144. Wainwright, D. A. et al. Durable therapeutic efficacy of murine gliomas. Clin. Cancer Res. 23, 124–136 inflammation by recruiting Hdac2 to specifically
utilizing combinatorial blockade against IDO, CTLA-4, (2017). repress IL-6. Nature 525, 389–393 (2015).
and PD-​L1 in mice with brain tumors. Clin. Cancer Res. 168. US National Library of Medicine. ClinicalTrials.gov, 191. Chheda, Z. S. et al. Novel and shared neoantigen
20, 5290–5301 (2014). https://ClinicalTrials.gov/show/NCT02658981 (2016). derived from histone 3 variant H3.3K27M mutation
145. Zeng, J. et al. Anti-​PD-1 blockade and stereotactic 169. US National Library of Medicine. ClinicalTrials.gov, for glioma T cell therapy. J. Exp. Med. 215, 141–157
radiation produce long-​term survival in mice with https://ClinicalTrials.gov/show/NCT02817633 (2016). (2018).

Nature Reviews | Cancer


Reviews

192. Mount, C. W. et al. Potent antitumor efficacy of anti-​ 206. Mitchell, D. A. et al. Tetanus toxoid and CCL3 improve 230. Farber, S. H. et al. Embracing rejection: immunologic
GD2 CAR T cells in H3-K27M+ diffuse midline gliomas. dendritic cell vaccines in mice and glioblastoma trends in brain metastasis. Oncoimmunology 5,
Nat. Med. 24, 572–579 (2018). patients. Nature 519, 366–369 (2015). e1172153 (2016).
193. Sampson, J. H. et al. Immunologic escape after This study demonstrates that the activation of 231. Fischer, G. M. et al. Molecular profiling reveals
prolonged progression-​free survival with epidermal memory T cells prior to DC vaccination leads to unique immune and metabolic features of melanoma
growth factor receptor variant III peptide vaccination increased migration of DCs to lymph nodes and brain metastases. Cancer Discov. 9, 628–645
in patients with newly diagnosed glioblastoma. J. Clin. improved clinical responses. (2019).
Oncol. 28, 4722–4729 (2010). 207. Garg, A. D., Coulie, P. G., Van den Eynde, B. J. & 232. Badie, B. & Schartner, J. M. Flow cytometric
194. Weller, M. et al. Rindopepimut with temozolomide for Agostinis, P. Integrating next-​generation dendritic characterization of tumor-​associated macrophages in
patients with newly diagnosed, EGFRvIII-​expressing cell vaccines into the current cancer immunotherapy experimental gliomas. Neurosurgery 46, 957–961;
glioblastoma (ACT IV): a randomised, double-​blind, landscape. Trends Immunol. 38, 577–593 (2017). discussion 961–962 (2000).
international phase 3 trial. Lancet Oncol. 18, 208. Nandu, H., Wen, P. Y. & Huang, R. Y. Imaging in 233. Bloch, O. et al. Gliomas promote immunosuppression
1373–1385 (2017). neuro-oncology. Ther. Adv. Neurol. Disord. 11, through induction of B7-H1 expression in tumor-​
195. Oji, Y. et al. Association of WT1 IgG antibody against 1756286418759865 (2018). associated macrophages. Clin. Cancer Res. 19,
WT1 peptide with prolonged survival in glioblastoma 209. Aquino, D., Gioppo, A., Finocchiaro, G., Bruzzone, M. G. 3165–3175 (2013).
multiforme patients vaccinated with WT1 peptide. & Cuccarini, V. MRI in glioma immunotherapy: evidence, 234. Parney, I. F., Waldron, J. S. & Parsa, A. T. Flow
Int. J. Cancer 139, 1391–1401 (2016). pitfalls, and perspectives. J. Immunol. Res. 2017, cytometry and in vitro analysis of human glioma-​
196. Rampling, R. et al. A cancer research UK first time in 5813951 (2017). associated macrophages: laboratory investigation.
human phase I Trial of IMA950 (novel multipeptide 210. Park, J. et al. Immune checkpoint inhibitor-​induced J. Neurosurg. 110, 572–582 (2009).
therapeutic vaccine) in patients with newly diagnosed reinvigoration of tumor-​infiltrating CD8+ T cells is 235. Sugihara, A. Q., Rolle, C. E. & Lesniak, M. S.
glioblastoma. Clin. Cancer Res. 22, 4776–4785 determined by their differentiation status in Regulatory T cells actively infiltrate metastatic brain
(2016). glioblastoma. Clin. Cancer Res. 25, 2549–2559 tumors. Int. J. Oncol. 34, 1533–1540 (2009).
197. Pollack, I. F. et al. Antigen-​specific immune responses (2019). 236. Berghoff, A. S. et al. Tumour-​infiltrating lymphocytes
and clinical outcome after vaccination with glioma-​ 211. US National Library of Medicine. ClinicalTrials.gov, and expression of programmed death ligand 1 (PD-​L1)
associated antigen peptides and polyinosinic– https://ClinicalTrials.gov/show/NCT03068832 (2017). in melanoma brain metastases. Histopathology 66,
polycytidylic acid stabilized by lysine and 212. US National Library of Medicine. ClinicalTrials.gov, 289–299 (2015).
carboxymethylcellulose in children with newly https://ClinicalTrials.gov/show/NCT04015700 (2019).
diagnosed malignant brainstem and nonbrainstem 213. US National Library of Medicine. ClinicalTrials.gov, Acknowledgements
gliomas. J. Clin. Oncol. 32, 2050–2058 (2014). https://ClinicalTrials.gov/show/NCT03422094 (2018). This work was supported by grants from the National Institutes
198. Liau, L. M. et al. Dendritic cell vaccination in 214. US National Library of Medicine. ClinicalTrials.gov, of Health to J.H.S.: R01-NS099463, P50-CA190991,
glioblastoma patients induces systemic and intracranial https://ClinicalTrials.gov/show/NCT03726515 (2018). P 3 0 -CA1 4 2 3 6 , P 01 -CA2 2 5 6 2 2 , R01 - CA 23 5 61 2 ,
T-​cell responses modulated by the local central nervous 215. US National Library of Medicine. ClinicalTrials.gov, U01-NS090284, R01-NS085412 and R01-CA175517. The
system tumor microenvironment. Clin. Cancer Res. 11, https://ClinicalTrials.gov/show/NCT03638167 (2018). funders had no role in the preparation of the manuscript or
5515–5525 (2005). 216. US National Library of Medicine. ClinicalTrials.gov, the decision to publish. The authors acknowledge T. Wright for
199. Prins, R. M. et al. Gene expression profile correlates https://ClinicalTrials.gov/show/NCT03500991 (2018). her assistance with manuscript preparation.
with T-​cell infiltration and relative survival in 217. US National Library of Medicine. ClinicalTrials.gov,
glioblastoma patients vaccinated with dendritic cell https://ClinicalTrials.gov/show/NCT02442297 (2015). Author contributions
immunotherapy. Clin. Cancer Res. 17, 1603–1615 218. US National Library of Medicine. ClinicalTrials.gov, All authors provided substantial contributions to the discus-
(2011). https://ClinicalTrials.gov/show/NCT02208362 (2014). sions of the content, made significant contributions to the
200. Okada, H. et al. Induction of CD8+ T-​cell responses 219. US National Library of Medicine. ClinicalTrials.gov, writing of the article and contributed equally to the review
against novel glioma-​associated antigen peptides and https://ClinicalTrials.gov/show/NCT03299309 (2017). and editing of the manuscript before submission.
clinical activity by vaccinations with α-​type 1 polarized 220. US National Library of Medicine. ClinicalTrials.gov,
dendritic cells and polyinosinic-​polycytidylic acid https://ClinicalTrials.gov/show/NCT02529072 (2015). Competing interests
stabilized by lysine and carboxymethylcellulose in 221. US National Library of Medicine. ClinicalTrials.gov, J.H.S. has an equity interest in Annias Immunotherapeutics,
patients with recurrent malignant glioma. J. Clin. Oncol. https://ClinicalTrials.gov/show/NCT02193347 (2014). which has licensed intellectual property from Duke related to
29, 330–336 (2011). 222. US National Library of Medicine. ClinicalTrials.gov, the use of the PEP-​CMV vaccine in the treatment of glioblas-
201. Phuphanich, S. et al. Phase I trial of a multi-​epitope- https://ClinicalTrials.gov/show/NCT03223103 (2017). toma. J.H.S. has an equity interest in Istari Oncology, which
pulsed dendritic cell vaccine for patients with newly 223. US National Library of Medicine. ClinicalTrials.gov, has licensed intellectual property from Duke related to the
diagnosed glioblastoma. Cancer Immunol. Immunother. https://ClinicalTrials.gov/show/NCT03914768 (2019). use of polio virus and D2C7 in the treatment of glioblastoma.
62, 125–135 (2013). 224. US National Library of Medicine. ClinicalTrials.gov, J.H.S. has additional relationships with Celldex (intellectual
202. Wen, P. Y. et al. A randomized, double-​blind, placebo-​ https://ClinicalTrials.gov/show/NCT03149003 (2017). property, royalties) and Medicenna Therapeutics (consulting).
controlled phase 2 trial of dendritic cell (DC) vaccination 225. US National Library of Medicine. ClinicalTrials.gov, J.H.S. is an inventor on patents related to the PEP-​CMV DC
with ICT-107 in newly diagnosed glioblastoma (GBM) https://ClinicalTrials.gov/show/NCT01204684 (2010). vaccine with tetanus, as well as to polio virus vaccine and
patients. J. Clin. Oncol. 32, 2005–2005 (2014). 226. US National Library of Medicine. ClinicalTrials.gov, D2C7 in the treatment of glioblastoma. D.M.A is an inventor
203. Liau, L. M. et al. First results on survival from a large https://ClinicalTrials.gov/show/NCT03927222 (2019). on patents related to the use of polio virus. M.D.G is an inven-
Phase 3 clinical trial of an autologous dendritic cell 227. US National Library of Medicine. ClinicalTrials.gov, tor on patents related to cellular vaccines. Duke and certain
vaccine in newly diagnosed glioblastoma. J. Transl. Med. https://ClinicalTrials.gov/show/NCT03688178 (2018). Duke investigators could benefit financially if related thera-
16, 142 (2018). 228. Peng, D. et al. Epigenetic silencing of TH1-type pies prove effective and are commercially successful. P.E.F.
204. Van Brussel, I., Berneman, Z. N. & Cools, N. Optimizing chemokines shapes tumour immunity and has no relevant disclosures.
dendritic cell-​based immunotherapy: tackling the immunotherapy. Nature 527, 249–253 (2015).
complexity of different arms of the immune system. 229. Johnson, L. A., Sanchez-​Perez, L., Suryadevara, C. M. Publisher’s note
Mediators Inflamm. 2012, 690643 (2012). & Sampson, J. H. Chimeric antigen receptor Springer Nature remains neutral with regard to jurisdictional
205. Mailliard, R. B. et al. Alpha-​type-1 polarized dendritic engineered T cells can eliminate brain tumors and claims in published maps and institutional affiliations.
cells: a novel immunization tool with optimized CTL-​ initiate long-​term protection against recurrence.
inducing activity. Cancer Res. 64, 5934–5937 (2004). Oncoimmunology 3, e944059 (2014). © Springer Nature Limited 2019

www.nature.com/nrc

You might also like