Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

J Neurooncol (2015) 123:425–432

DOI 10.1007/s11060-015-1830-1

EDITORS’ INVITED MANUSCRIPT

Dendritic cell immunotherapy for brain tumors


Joseph P. Antonios1 • Richard G. Everson1 • Linda M. Liau1,2,3,4

Received: 2 April 2015 / Accepted: 25 May 2015 / Published online: 3 June 2015
Ó Springer Science+Business Media New York 2015

Abstract Glioblastomas are characterized by immuno- Introduction


suppression, rapid proliferation, angiogenesis, and invasion
into the surrounding brain parenchyma. Limitations in Malignant gliomas are the most frequent and aggressive
current therapeutic approaches have spurred the develop- intrinsic brain tumors of the central nervous system (CNS).
ment of personalized, patient-specific treatments. Among Characterized by diffuse infiltration into brain parenchyma,
these, active immunotherapy has emerged as a viable gliomas often do not respond well to conventional treatments
option for glioma treatment. The ability to generate an [1, 2]. There is an urgent need for more effective and specific
immune response utilizing patient-derived dendritic cells therapies, as current treatment methods of surgery,
(DCs) (professional antigen-presenting cells) is especially chemotherapy and radiation have failed to substantially alter the
attractive. This approach to glioma treatment allows for the poor prognosis of this disease [1, 3]. Immunotherapeutic
immunologic targeting and destruction of malignant cells. approaches to glioma treatment are especially attractive, as they
Data acquired in multiple pre-clinical models and clinical allow for specific targeting and destruction of malignant cells.
trials have shown significant responses and prolonged Indeed, while the CNS was once considered an immune-priv-
survival. Here we provide an overview of the current status ileged site largely protected by the blood–brain barrier, it has
of DC vaccination for the treatment of gliomas. been shown that, instead, the CNS supports a highly-specialized
immune surveillance system that can recruit cells associated
Keywords Glioma  Glioblastoma  Immunotherapy  with both innate and adaptive immunity [4]. Specifically, den-
Dendritic cell vaccine dritic cell (DC) immunotherapy is an attractive approach for the
treatment of brain tumors, as immune cells theoretically can
seek out and selectively destroy invading malignant cells while
sparing normal brain tissues [3]. As professional antigen pre-
& Linda M. Liau senting cells (APCs), DCs are the most potent stimulators of the
lliau@mednet.ucla.edu
cell-mediated immune system. Its specialized ability to
1
Department of Neurosurgery, David Geffen School of orchestrate antigen-specific responses through CD4? helper T
Medicine at UCLA, University of California Los Angeles, cells and CD8? cytotoxic T cells makes DCs prime candidates
Los Angeles, CA 90095, USA to induce anti-tumor immunity. This review discusses the pro-
2
Brain Research Institute, David Geffen School of Medicine at mise these DC-based immunotherapies have shown for treating
UCLA, University of California Los Angeles, Los Angeles, malignant brain tumors.
CA 90095, USA
3
Jonsson Comprehensive Cancer Center, David Geffen School
of Medicine at UCLA, University of California Los Angeles,
Los Angeles, CA 90095, USA
Dendritic cells and cancer immunotherapy
4
Department of Neurosurgery, David Geffen School of
DCs are the foremost coordinators of immune responses,
Medicine at UCLA, University of California Los Angeles,
300 Stein Plaza, Suite 562, Box 956901, Los Angeles, bridging non-antigen specific innate immunity with the
CA 90095-6901, USA antigen-specific arm of adaptive immunity [5]. Though

123
426 J Neurooncol (2015) 123:425–432

DCs are most often known as professional APCs, their DCs involves culturing peripheral monocytes with the
immature state is characterized by high levels of phago- cytokines interleukin-4 (IL-4) and granulocyte–macro-
cytic activity and not antigen presentation [6]. They are phage colony-stimulating factor [8]. The resulting pheno-
localized in most organs and tissues, especially where type of these cells most closely resembles conventional
pathogen entry is most likely, and act as local sentinels and DCs, which are characterized by high levels of CD11c and
primary immune responders. By constantly sampling and MHC Class II expression and can stimulate T cell prolif-
processing antigens, DCs make the crucial decision to eration. The resulting immature DCs have proven to be
induce tolerance or begin the recruitment of effector cells. fully effective in antigen uptake and subsequent T cell
Once they have trafficked into draining lymph nodes, activation once mature. Generation of immature DCs is
mature DCs can quickly and efficiently seek out rare necessary for most methods of vaccine preparation. This
antigen-specific T cells to begin a proliferative burst stems from the enhanced phagocytic abilities of immature
(Fig. 1) [7]. Their specialized ability to orchestrate antigen- DCs compared to their mature counterparts, which allows
specific responses through CD4? helper T cells and CD8? for the uptake of antigen sources [9]. Immature DCs can be
cytotoxic T cells makes DCs prime candidates to induce pulsed with autologous tumor-eluted peptides [10], tumor
anti-tumor immunity. lysate [11], or tumor-derived RNA [12]. These must then
With respect to cancer, in the process of malignant be matured and activated—a necessary step for upregula-
transformation and mutation, many neoplastic cells give tion of co-stimulatory molecules, antigen presentation, and
rise to proteins that are not normally expressed in most chemokine receptors associated with migration. DCs can
tissues of the body. The presence of unique, identifying be administered intradermally for maturation in vivo; or
antigens renders tumor cells susceptible to immune they can be matured ex vivo with the addition of adjuvants
recognition. When immune regulation remains competent, (e.g., a-galactosylceramide, CD40-specific antibody,
tumor cells express these unique antigens, allowing circu- inflammatory cytokines, or specific TLR ligands such as
lating DCs to recognize them as foreign. Consequently, DC polyI:C) and then pulsed with defined T cell epitopes
vaccination has proven to be promising in treating various [13].More recently, Kalinski et al. developed mature, non-
types of cancers, including human gliomas. exhausted ‘type-1-polarized’ DCs (DC1s) that produce
DCs originate in the bone marrow from precursor stem high levels of IL-12 by exposing immature DCs to types I
cells. The most common in vitro method used to generate and II interferons in culture [14]. Okada et al. showed that

Fig. 1 Dendritic cells originate in the bone marrow. Once they enter will mature and seek out an antigen-specific T cell to begin a
tissues and organs, the immature dendritic cells sample and process proliferative burst. HSC hematopoietic stem cell, CMP common
antigens from the environment. A dendritic cell that has processed myeloid progenitor, MDP macrophage dendritic cell progenitor, CDP
antigen will partially mature and migrate to a lymph node, where it common dendritic cell progenitor

123
J Neurooncol (2015) 123:425–432 427

these novel DC1-type DCs induced long-lived T-cell escalation trial, 12 malignant glioma patients were treated
responses against targeted antigens more efficiently than with DCs loaded with acid-eluted peptides from autologous
mature DCs [15]. tumor. This DC immunotherapy was found to be safe and
well-tolerated, as no cases of either autoimmunity or dose-
limiting toxicity were reported with up to 107 DCs injected
DC-based immunotherapy for brain tumors per dose [10]. Patients in this study exhibited increased
progression-free survival and median overall survival
Developing clinically relevant DC vaccine therapies has compared to historical controls, with five long-term sur-
proven challenging, with multiple approaches of preparing vivors ([36 months). Of the eight patients who developed
DCs for vaccination tested in clinical trials for patients with new areas of contrast enhancement on follow-up MRI
gliomas (Table 1). scans, four demonstrated prolonged survival following
surgical biopsy and resection. Specimens obtained from
Acid-eluted peptide DC vaccine these patients demonstrated a significant CD3? T-cell
infiltrate, predominantly composed of memory T cells
Liau et al. described the first case report of a glioblastoma (CD8?/CD45RO?). Data from these initial trials also
patient treated with DC-based immunotherapy, which suggested that DC-based immunotherapy works best in the
consisted of three intradermal injections of autologous DCs absence of actively progressive tumor and with minimal
pulsed with allogeneic acid-eluted peptides [16]. Due to the residual disease.
marked heterogeneity of malignant gliomas, this was an Subsequently, Yu et al. also reported on glioblastoma
attractive approach to DC therapy [17]. The acid-elution patients treated with intradermal DCs loaded with acid-
technique extracted tumor antigens bound to MHC Class I eluted tumor peptides with similar findings [18]. Never-
molecules, allowing for the preparation of a unique tumor- theless, this approach specifically utilizing DCs prepared
antigen specific vaccine. While clinical benefit was diffi- with acid-eluted peptides was somewhat limited. This was
cult to assess in this single case report, an increased CD3? largely due to technical difficulties related to the require-
T-cell infiltrate into the tumor was noted. In a Phase I dose- ment for large numbers of primary tumor cells needed for

Table 1 Clinical trials assessing dendritic cell immunotherapy


Therapy/protocol Center/principal investigator Phase- ND, P, R WHO ClinicalTrials.gov References
enrollment (newly grades identifier
diagnosed,
persistent,
recurrent)

Acid-eluted tumor peptide UCLA, Los Angeles, CA/Liau I-28 ND III or IV NCT00068510 [14]
dendritic cell vaccine
Autologous tumor lysate UCLA, Los Angeles, CA/Liau II-23 ND, R III or IV NCT01204684 [11, 15]
dendritic cell vaccine
Autologous tumor lysate UCLA, Los Angeles, CA/Liau III-348 ND IV NCT00045968
dendritic cell vaccine
Autologous tumor lysate Cedars-Sinai Medical Center, Los II-50 R III or IV NCT00576537 [17, 19]
dendritic cell vaccine Angeles, CA/Yu
Autologous tumor lysate University Hospital Gasthuisberg, 56 R IV [20]
dendritic cell vaccine Leuven, Belgium/van Gool
Autologous tumor lysate University Hospital Gasthuisberg, 45 ND III or IV [21]
dendritic cell vaccine Leuven, Belgium/van Gool
Tumor-specific EGFRvIII Duke University, Durham, NC and II-40 ND IV NCT00643097 [23]
vaccine MD Anderson Cancer Center at
UT Houston, Houston, TX/
Vlahovic and Sampson
Autologous DC ? brain tumor Duke University, Durham, NC/ I-50 R IV NCT00890032
stem cell-mRNA Sampson
Autologous DCs cancer stem Huashan Hospital, Shanghai, II ND, R III or IV NCT01567202
cell vaccine China/Zhou
Table categorized based on therapy and protocol, with associated information regarding phase, grade, clinical trial identifier, and any associated
references

123
428 J Neurooncol (2015) 123:425–432

acid-elution and the inability to consistently grow out booster vaccines of tumor lysate alone. Patients on the trial
sufficient tumor cells for this purpose. had a median survival of 9.6 months, with some patients
surviving more than 3 years from diagnosis of recurrent
Tumor-lysate pulsed DC vaccine glioblastoma. Patients who underwent complete resection
of tumor demonstrated a statistically significant improve-
More recently, tumor lysate prepared from autologous ment in survival versus those who did not. A small per-
tumor cells obtained from surgical resection have been centage of patients in this trial displayed transient
utilized to pulse DCs. In this technique, patient tumor cells worsening of focal neurologic deficits that had typically not
are lysed using freeze/thaw cycles to generate cellular been reported in other trials. Whether or not these patients
fragments; and exposure of DCs to these tumor fragments had residual tumor was not reported. Nine of 21 patients on
allows for phagocytosis and subsequent peptide antigen this trial demonstrated a positive delayed-type hypersen-
presentation on both MHC Classes I and II. Although there sitivity (DTH) response. However, DTH responses did not
is a theoretical disadvantage of the increased potential for correlate with overall survival. Ardon et al. expanded their
selection of auto-reactive T cells due to the lack of antigen study to evaluate the efficacy of DC vaccination in pedi-
specificity, no severe auto-immunity has ever been reported atric patients who presented with a variety of high-grade
with the use of tumor lysate-pulsed DC vaccination for malignant brain tumors [23]. Autologous tumor lysate was
CNS tumors. used as the antigen source for the autologous monocyte-
Prins et al. reported on a Phase I/II clinical trial that derived DC vaccine. Of those in the study, patients with
involved patients with both newly diagnosed and recurrent high-grade glioma and atypical teratoid–rhabdoid tumors
glioblastoma receiving immunotherapy with autologous seemed to respond favorably to treatment, whereas patients
tumor lysate-pulsed DCs, as well as adjuvant imiquimod or with ependymoma or medulloblastoma/PNETs did not.
poly-ICLC during booster vaccinations. Safety of the vac- Similar to their adult trials, this group observed a subset of
cines and the adjuvants were demonstrated, with no sig- patients diagnosed with high-grade gliomas exhibited long-
nificant adverse events reported. Patients in this trial had term survival.
substantially better progression-free and overall survival
rates compared to historical controls, with a median sur- Glioma-associated antigen pulsed DC vaccine
vival of 31.4 months for newly diagnosed glioblastoma
patients. Improved outcome appeared also to stratify by Several glioma-associated peptides have also been used to
tumor molecular subtype as measured by gene expression pulse DCs for clinical trials in malignant glioma patients.
profiling, with increased survival shown for patients within These tumor-associated antigens (TAAs) have included the
the mesenchymal subgroup of glioblastoma [11]. Currently, mutated form of the epidermal growth factor receptor
a Phase III multi-center trial of DC vaccination for newly (EGFRvIII), as well as the IL-13 receptor a2 (IL-13a2) and
diagnosed glioblastoma is underway, with approximately gp100 [15, 24]. These peptides were able to induce specific
348 patients to be enrolled (NCT00045968) [19]. responses in patients, as measured by in vitro proliferation
Yu et al. also reported a Phase II trial with DC assays and measurement of tetramer positive CD8? T cells.
immunotherapy in patients with recurrent glioblastoma that Sampson et al. targeted EGFRvIII with DCs pulsed with a
utilized DCs loaded with autologous tumor lysate [20]. They peptide spanning the fusion junction of EGFRvIII conju-
found evidence of systemic antitumor CTL activity, as well gated to KLH [24, 25]. EGFRvIII expression in the tumor
as intra-tumoral lymphocyte infiltration in half of patients was not an eligibility criterion for entry into the trial which
undergoing re-operation. Median survival in the DC vacci- was a Phase I toxicity study. They intended to treat 15
nated patients was markedly increased to 133 versus patients, 3 of which progressed prior to receiving the
30 weeks for matched historical control patients treated at vaccine. Of the vaccinated patients, many displayed
the same institution. Wheeler et al. reported that after vac- EGFRvIII-specific immune responses with 10 out of 12
cination, PBMCs restimulated by tumor lysate were found to demonstrating antigen-specific T-cell proliferation and 5
increase production of IFN-c by greater than 1.5-fold in a out of 9 showing DTH responses to EGFRvIII.
significant percentage of patients on the trial [21]. These DCs loaded with tumor-derived mRNA or cancer stem
patients, who were characterized as ‘‘vaccine responders,’’ cell antigens have also been studied [26]. An ongoing
demonstrated an increased survival compared to ‘‘non-re- Phase I trial is currently examining the applicability of a
sponders’’. Furthermore, vaccine response appeared to vaccine utilizing mRNA-loaded DCs (NCT00890032) for
potentiate the effects of subsequent chemotherapy. patients with gliomas. A Phase II trial is alternatively
De Vleeschouwer et al. reported on the treatment 56 examining DCs loaded with glioma stem-like cell antigens
patients with recurrent glioblastoma with matured DCs in (NCT01567202). Although these studies have not demon-
varied vaccination schedules [22]. They also included strated significantly increased overall survival, their ability

123
J Neurooncol (2015) 123:425–432 429

to induce antigen specific responses may warrant further I interferons, IFN-a, IFN-b and IFN-x, are produced in the
investigation. setting of viral antigens and immune-modulatory signaling.
IFN-a induces tumor cell apoptosis and stimulates DCs. At
this point, however, clinical studies have not shown sig-
Vaccine adjuvants nificant benefit with the adjuvant use of IFN-a [30]. Type II
interferons, such as IFN-c, are released from activated T
The ultimate goal of DC vaccine immunotherapy is to cells to promote anti-tumor activity [31]. In pre-clinical
induce a long-term immune response in glioma patients. studies, administration of IFN-c alongside glioma-lysate
Gliomas express cytokines [such as transforming growth vaccination promoted anti-tumor activity with a significant
factor-b2 (TGF-b2) and IL-10] that are known to sup- increase in long-term survival [32].
press immune responses both directly and indirectly Additionally, ILs—another family of cytokines—regu-
(through T regulatory cells; Fig. 2). To promote a suc- late immune response. IL-2 promotes T-cell proliferation
cessful long-term anti-tumor response and counter and function. Therapies using IL-2 alone in pre-clinical
immunosuppressive factors, the use of adjuvant treatments tumor-bearing animal models prolonged survival, with
alongside DC vaccine to counter tumor immunosuppres- some promise shown in patients when used as an adjuvant
sion has been extensively studied [27]. therapy [33].Systemic IL-4, produced by T cells, promotes
Initially hampered by unfavorable side effects, pre- a Th2 response [34]. It has been shown, however, that local
clinical work has revived the use of immunotherapeutic delivery of IL-4 at the site of immunization induces IL-12
adjuvants, with the advent of engineered local delivery production [35]. IL-12 promotes the Th1 response, as well
systems with lower associated toxicities [28, 29]. as T-cell activation and IFN-c production. Similarly, pre-
Interferons, which activate and mediate immune clinical models have shown a survival benefit with
response, are divided into two classes: types II and I. Type administration of this cytokine both alone and as an adju-
vant to other therapeutic approaches [36].
Recently, Mitchell et al. demonstrated that using a recall
antigen to pre-condition the vaccine site allowed for
improved vaccination efficiency. They demonstrated
improved clinical responses in glioblastoma patients via this
technique; specifically, skin that was preconditioned with
tetanus/diphtheria toxoid stimulated increased CCL3 pro-
duction, in turn allowing DCs to better traffic into the local
lymph nodes and promote better T cell responses [37].

DC immunotherapy and checkpoint inhibitors

The immune system is intrinsically primed to respond to


infectious agents and harmful substances. From recogniz-
ing, containing, and eliminating pathogens to developing
immunologic memory and responding to re-exposure, the
immune system is able to react to most biologic insults. In
the case of tumor, however, several mechanisms come into
play. The first line of anti-tumor defense, innate immunity,
relies on phagocytic cells, granulocytes, and lymphoid
progenitor-derived natural killer cells to mediate an initial
immune response. DC activation and migration to lymphoid
tissue upon phagocytosis of malignant, dying, or stressed
cells then leads to activation of secondary immune response.
This adaptive immunity is mediated by the presentation of
TAAs by APCs to induce TAA-dependent clonal expansion
of T cells (CD8?) cytotoxic T cells (CTLs), CD4? helper T
Fig. 2 Glioma tumor cells release cytokines that promote a suppres-
cells, and regulatory T lymphocytes (Tregs) [38].
sive environment by both directly inhibiting the anti-tumor response
and promoting further release of inhibitory factors from cells such as Several factors may limit the ability of the immune
T regulatory cells system to mount a successful adaptive response against

123
430 J Neurooncol (2015) 123:425–432

gliomas (Fig. 3). Relevant to this discussion, cytotoxic These findings suggest that gliomas are able to success-
T-lymphocyte associated antigen 4 (CTLA-4) and the fully alter the tumor microenvironment such that effective
programmed death 1 (PD-1) receptor have both been anti-tumor immune responses are suppressed. Indeed,
identified as negative immune regulators [39]. CTLA-4 is a clinical studies inhibiting these suppressive mechanisms
co-inhibitory receptor expressed on immune cells that have been pioneered and suggested therapeutic benefit in
competes with CD28? receptor binding of CD80 and CD86 melanoma [45]. Pre-clinical glioma models have corrobo-
to inhibit the initial activation of T cells [40]. Recent rated this evidence, with Wainwright et al. demonstrating
studies similarly show that the inhibition of CTLA-4 by that blockade of CTLA-4, PD-1, and IDO in established
antibody blockade results in objective tumor regression in intracranial glioma models resulted in decreased tumor-in-
patients with metastatic melanoma [41, 42]. filtrating T regulatory cells and increased long-term sur-
Glioma cells themselves reduce T-lymphocyte prolifer- vival mediated by cytotoxic T lymphocytes [47]. Similarly,
ative function, decrease Th1 cytokine synthesis, limit Zeng et al. showed that PD-1 blockade in the setting of
interactions with normal cells, and alter HLA Class I radiation therapy led to long-term survival in mice
antigen expression [43]. Expression of TGF-b inhibits implanted with a murine glioma cell line [46]. These results
T-cell proliferation and CTL development, as well pro- suggest promise for the inhibition of these immune-regu-
motes tumor vascularization and glioma cell migration latory mechanisms for the potentiation of immune response
[44]. PD-1, an immune regulatory receptor associated with induced by DC-based vaccination strategies.
inhibition of T cells with prolonged antigen exposure (a
phenomenon noted in chronic disease manifestations, such
as viral infections and cancer), plays an important role in Conclusion
regulating the effector phase of the T-cell response [45].
Activation of this receptor by PD-L1 and PD-L2, expressed In conclusion, DC vaccines have shown efficacy in multi-
by glioma cells and regulatory cells in the tumor ple pre-clinical studies and Phase I/II clinical trials for
microenvironment, promotes immune suppression [39, 41, gliomas. While acid-eluted peptide and glioma-associated
42]. Early studies have shown that inhibition of the PD-1 peptide preparations have shown some clinical benefit, the
receptor is associated with a significantly increased most widespread progress has been made using autologous
immune response to cancer [41, 46]. tumor lysate-pulsed DC vaccine approaches. Although it
was initially hypothesized that non-specific lysate prepa-
rations could induce severe autoimmune reactions in the
CNS, no major adverse autoimmune events have been
reported with now over 600 brain tumor patients treated to
date worldwide. Additionally, these vaccines have been
shown to induce antigen-specific responses, potentiate
immune cell infiltration into the tumor, and increase overall
survival in a subset of patients. Further developments in
adjuvant therapies will allow us to modulate and fine-tune
these anti-tumor immune responses. In the future, further
genomic sequencing and molecular characterizations of
brain tumors will undoubtedly allow for personalized DC
vaccines for glioblastoma patients that should generate
more potent and long-lasting anti-tumor immunity.

References

1. Stupp R, Mason WP, van den Bent MJ, Weller M, Fisher B,


Taphoorn MJ et al (2005) Radiotherapy plus concomitant and
adjuvant temozolomide for glioblastoma. N Engl J Med
352:987–996
2. Hanahan D, Weinberg RA (2011) Hallmarks of cancer: the next
generation. Cell 144:646–674
Fig. 3 Mechanisms of inhibition in the tumor environment exist to 3. Yamanaka R, Kajiwara K (2012) Dendritic cell vaccines. Adv
restrict anti-glioma immune response Exp Med Biol 746:187–200

123
J Neurooncol (2015) 123:425–432 431

4. McLendon R, Friedman A, Bigner D, Van Meir EG, Brat DJ, 22. De Vleeschouwer S, Fieuws S, Rutkowski S, Van Calenbergh F,
Mastrogianakis GM et al (2008) Comprehensive genomic char- Van Loon J, Goffin J et al (2008) Postoperative adjuvant dendritic
acterization defines human glioblastoma genes and core path- cell-based immunotherapy in patients with relapsed glioblastoma
ways. Nature 455:1061–1068 multiforme. Clin Cancer Res 14:3098–3104
5. Trumpfheller C, Longhi MP, Caskey M, Idoyaga J, Bozzacco L, 23. Ardon H, De Vleeschouwer S, Van Calenbergh F, Claes L,
Keler T et al (2012) Dendritic cell-targeted protein vaccines: a Kramm CM, Rutkowski S et al (2010) Adjuvant dendritic cell-
novel approach to induce T-cell immunity. J Intern Med based tumour vaccination for children with malignant brain
271:183–192 tumours. Pediatr Blood Cancer 54:519–525
6. Steinman RM, Idoyaga J (2010) Features of the dendritic cell 24. Sampson JH, Archer GE, Mitchell DA, Heimberger AB, Herndon
lineage. Immunol Rev 234:5–17 JE II, Lally-Goss D et al (2009) An epidermal growth factor
7. Steinman RM, Banchereau J (2007) Taking dendritic cells into receptor variant III-targeted vaccine is safe and immunogenic in
medicine. Nature 449:419–426 patients with glioblastoma multiforme. Mol Cancer Ther
8. Shurin MR, Salter RD (2009) Dendritic cells in cancer. Springer, 8:2773–2779
New York 25. Morgan RA, Johnson LA, Davis JL, Zheng Z, Woolard KD, Reap
9. Liu K, Nussenzweig MC (2010) Origin and development of EA et al (2012) Recognition of glioma stem cells by genetically
dendritic cells. Immunol Rev 234:45–54 modified T cells targeting EGFRvIII and development of adop-
10. Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, tive cell therapy for glioma. Hum Gene Ther 23:1043–1053
Giovannone AJ et al (2005) Dendritic cell vaccination in 26. Cao JX, Zhang XY, Liu JL, Li D, Li JL, Liu YS et al (2014)
glioblastoma patients induces systemic and intracranial T-cell Clinical efficacy of tumor antigen-pulsed DC treatment for high-
responses modulated by the local central nervous system tumor grade glioma patients: evidence from a meta-analysis. PLoS ONE
microenvironment. Clin Cancer Res 11:5515–5525 9:e107173
11. Prins RM, Soto H, Konkankit V, Odesa SK, Eskin A, Yong WH 27. Butowski N (2010) Immunostimulants for malignant gliomas.
et al (2011) Gene expression profile correlates with T-cell infil- Neurosurg Clin N Am 21:53–65
tration and relative survival in glioblastoma patients vaccinated 28. Kamimura K, Suda T, Zhang G, Liu D (2011) Advances in gene
with dendritic cell immunotherapy. Clin Cancer Res 17:1603–1615 delivery systems. Pharm Med 25:293–306
12. Caruso DA, Orme LM, Neale AM, Radcliff FJ, Amor GM, 29. Rainov NG, Heidecke V (2011) Clinical development of exper-
Maixner W et al (2004) Results of a phase 1 study utilizing imental virus-mediated gene therapy for malignant glioma. Anti-
monocyte-derived dendritic cells pulsed with tumor RNA in cancer Agents Med Chem (Former Curr Med Chem Anti-cancer
children and young adults with brain cancer. Neuro-oncology Agents) 11:739–747
6:236–246 30. Chiocca EA, Smith KM, McKinney B, Palmer CA, Rosenfeld S,
13. Tacken PJ, De Vries IJM, Torensma R, Figdor CG (2007) Den- Lillehei K et al (2008) A phase I trial of Ad.hIFN-beta gene
dritic-cell immunotherapy: from ex vivo loading to in vivo tar- therapy for glioma. Mol Ther 16:618–626
geting. Nat Rev Immunol 7:790–802 31. Hervas-Stubbs S, Perez-Gracia JL, Rouzaut A, Sanmamed MF,
14. Kalinski P, Okada H (2010) Polarized dendritic cells as cancer Le Bon A, Melero I (2011) Direct effects of type I interferons on
vaccines: directing effector-type T cells to tumors. Semin cells of the immune system. Clin Cancer Res 17:2619–2627
Immunol 22(3):173–182 32. Pluhar GE, Grogan PT, Seiler C, Goulart M, SantaCruz KS,
15. Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE Carlson C et al (2010) Anti-tumor immune response correlates
et al (2011) Induction of CD8? T-cell responses against novel with neurological symptoms in a dog with spontaneous astrocy-
glioma-associated antigen peptides and clinical activity by vaccina- toma treated by gene and vaccine therapy. Vaccine 28:3371–3378
tions with alpha-type 1 polarized dendritic cells and polyinosinic– 33. Colombo F, Barzon L, Franchin E, Pacenti M, Pinna V, Danieli D
polycytidylic acid stabilized by lysine and carboxymethylcellulose in et al (2005) Combined HSV-TK/IL-2 gene therapy in patients
patients with recurrent malignant glioma. J Clin Oncol 29:330–336 with recurrent glioblastoma multiforme: biological and clinical
16. Liau LM, Black KL, Martin NA, Sykes SN, Bronstein JM, Jou- results. Cancer Gene Ther 12:835–848
ben-Steele L et al (2000) Treatment of a patient by vaccination 34. Serre K, Mohr E, Gaspal F, Lane PJ, Bird R, Cunningham AF
with autologous dendritic cells pulsed with allogeneic major et al (2010) IL-4 directs both CD4 and CD8 T cells to produce
histocompatibility complex class I-matched tumor peptides. Case Th2 cytokines in vitro, but only CD4 T cells produce these
Report. Neurosurg Focus 9:e8 cytokines in response to alum-precipitated protein in vivo. Mol
17. Prins RM, Wang X, Soto H, Young E, Lisiero DN, Fong B et al Immunol 47:1914–1922
(2013) Comparison of glioma-associated antigen peptide-loaded 35. Okada H, Lieberman FS, Walter KA, Lunsford LD, Kondziolka
versus autologous tumor lysate-loaded dendritic cell vaccination DS, Bejjani GK et al (2007) Autologous glioma cell vaccine
in malignant glioma patients. J Immunother 36:152–157 admixed with interleukin-4 gene transfected fibroblasts in the
18. Yu JS, Wheeler CJ, Zeltzer PM, Ying H, Finger DN, Lee PK et al treatment of patients with malignant gliomas. J Transl Med 5:67
(2001) Vaccination of malignant glioma patients with peptide- 36. Sonabend AM, Velicu S, Ulasov IV, Han Y, Tyler B, Brem H
pulsed dendritic cells elicits systemic cytotoxicity and intracra- et al (2008) A safety and efficacy study of local delivery of
nial T-cell infiltration. Cancer Res 61:842–847 interleukin-12 transgene by PPC polymer in a model of experi-
19. Polyzoidis S, Ashkan K (2014) DCVax(R)-L-developed by North- mental glioma. Anticancer Drugs 19:133–142
west Biotherapeutics. Hum Vaccine Immunother 10:3139–3145 37. Mitchell DA, Batich KA, Gunn MD, Huang M-N, Sanchez-Perez
20. Jouanneau E, Black KL, Veiga L, Cordner R, Goverdhana S, Zhai L, Nair SK et al (2015) Tetanus toxoid and CCL3 improve
Y et al (2014) Intrinsically de-sialylated CD103(?) CD8 T cells dendritic cell vaccines in mice and glioblastoma patients. Nature
mediate beneficial anti-glioma immune responses. Cancer 519:366–369
Immunol Immunother 63:911–924 38. Zou W (2006) Regulatory T cells, tumour immunity and
21. Wheeler CJ, Black KL, Liu G, Mazer M, Zhang XX, Pepkowitz S immunotherapy. Nat Rev Immunol 6:295–307
et al (2008) Vaccination elicits correlated immune and clinical 39. Ott PA, Hodi FS, Robert C (2013) CTLA-4 and PD-1/PD-L1
responses in glioblastoma multiforme patients. Cancer Res blockade: new immunotherapeutic modalities with durable clin-
68:5955–5964 ical benefit in melanoma patients. Clin Cancer Res 19:5300–5309

123
432 J Neurooncol (2015) 123:425–432

40. Fong B, Jin R, Wang X, Safaee M, Lisiero DN, Yang I et al 44. Grauer O, Pschl P, Lohmeier A, Adema GJ, Bogdahn U (2007)
(2012) Monitoring of regulatory T cell frequencies and expres- Toll-like receptor triggered dendritic cell maturation and IL-12
sion of CTLA-4 on T cells, before and after DC vaccination, can secretion are necessary to overcome T-cell inhibition by glioma-
predict survival in GBM patients. PLoS ONE 7:e32614 associated TGF-beta2. J Neurooncol 82:151–161
41. Hamid O, Robert C, Daud A, Hodi FS, Hwu W-J, Kefford R et al 45. Ribas A (2012) Tumor immunotherapy directed at PD-1. N Engl
(2013) Safety and tumor responses with lambrolizumab (anti-PD- J Med 366:2517–2519
1) in melanoma. N Engl J Med 369:134–144 46. Zeng J, See AP, Phallen J, Jackson CM, Belcaid Z, Ruzevick J
42. Curran MA, Montalvo W, Yagita H, Allison JP (2010) PD-1 and et al (2013) Anti-PD-1 blockade and stereotactic radiation pro-
CTLA-4 combination blockade expands infiltrating T cells and duce long-term survival in mice with intracranial gliomas. Int J
reduces regulatory T and myeloid cells within B16 melanoma Radiat Oncol Biol Phys 86:343–349
tumors. Proc Natl Acad Sci USA 107:4275–4280 47. Wainwright DA, Chang AL, Dey M, Balyasnikova IV, Kim C,
43. Schartner JM, Hagar AR, Van Handel M, Zhang L, Nadkarni N, Tobias AL et al (2014) Durable therapeutic efficacy utilizing
Badie B (2005) Impaired capacity for upregulation of MHC class combinatorial blockade against IDO, CTLA-4 and PD-L1 in mice
II in tumor-associated microglia. Glia 51:279–285 with brain tumors. Clin Cancer Res 20(20):5290–5301

123

You might also like