Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2017 Dec 1.
Published in final edited form as: J Neurooncol. 2016 Sep 13;130(3):517–527. doi: 10.1007/s11060-016-2245-3

Antigen-specific immunoreactivity and clinical outcome following vaccination with glioma-associated antigen peptides in children with recurrent high-grade gliomas: Results of a pilot study

Ian F Pollack 1,8,9, Regina I Jakacki 2,8,9, Lisa H Butterfield 4,5,7,9, Ronald L Hamilton 3,9, Ashok Panigrahy 6,8, Daniel P Normolle 10, Angela K Connelly 2,8, Sharon Dibridge 2,8, Gary Mason 2,8,9, Theresa L Whiteside 3,7,9, Hideho Okada 1,5,7,9,11
PMCID: PMC5363717  NIHMSID: NIHMS852467  PMID: 27624914

Abstract

Purpose

Recurrent high-grade gliomas (HGGs) of childhood have an exceedingly poor prognosis with current therapies. Accordingly, new treatment approaches are needed. We initiated a pilot trial of vaccinations with peptide epitopes derived from glioma-associated antigens (GAAs) overexpressed in these tumors in HLA-A2+ children with recurrent HGG that had progressed after prior treatments.

Methods

Peptide epitopes for three GAAs (EphA2, IL13Rα2, survivin), emulsified in Montanide-ISA-51, were administered subcutaneously adjacent to intramuscular injections of poly-ICLC every 3 weeks for 8 courses, followed by booster vaccines every six weeks. Primary endpoints were safety and T-cell responses against the GAA epitopes, assessed by enzyme-linked immunosorbent spot (ELISPOT) analysis. Treatment response was evaluated clinically and by magnetic resonance imaging.

Results

Twelve children were enrolled, 6 with glioblastoma, 5 with anaplastic astrocytoma, and one with malignant gliomatosis cerebri. No dose-limiting non-CNS toxicity was encountered. ELISPOT analysis, in ten children, showed GAA responses in 9: to IL13Rα2 in 4, EphA2 in 9, and survivin in 3. One child had presumed symptomatic pseudoprogression, discontinued vaccine therapy, and responded to subsequent treatment. One other child had a partial response that persisted throughout two years of vaccine therapy, and continues at > 39 months. Median progression-free survival (PFS) from the start of vaccination was 4.1 months and median overall survival (OS) was 12.9 months. Six-month PFS and OS were 33% and 73%, respectively.

Conclusion

GAA peptide vaccination in children with recurrent malignant gliomas is generally well tolerated, and has preliminary evidence of immunological and modest clinical activity.

Keywords: astrocytoma, glioma, immunotherapy, pediatric brain tumor, vaccine therapy

INTRODUCTION

Children with malignant gliomas have high rates of disease progression after initial therapy with irradiation and adjuvant chemotherapy, with five-year survival rates less than 20%[13]. After progression, the likelihood of prolonged survival is low, with poor response rates to numerous conventional[4, 5] and molecularly targeted[6, 7] agents.

Accordingly, there is a need to identify new therapeutic approaches to promote tumor cell killing by targeting distinctive features of tumor cells. Cancer vaccines, designed to induce systemic immunity against antigens overexpressed by tumor cells, are promising in this regard. Pilot clinical trials by us [812] and others [1318] have demonstrated the safety and potential efficacy of peripheral vaccinations for adults with malignant gliomas. Vaccine approaches may be even more effective if applied in clinical scenarios where patients are likely to have intact immunity, such as the pediatric age group,[1921] as we have recently demonstrated in a pilot trial for children with newly diagnosed brainstem gliomas.[22]

The current trial incorporated peptide epitopes for three glioma-associated antigens (GAAs) that we demonstrated were highly expressed in pediatric gliomas, IL13Rα2, EphA2, and survivin.[23] The human leukocyte antigen (HLA)-A2-restricted cytotoxic T-lymphocyte (CTL) epitopes included an interleukin-13 receptor (IL-13Rα2) analog peptide (IL-13Rα2345–353:1A9V)[24, 25] and EphA2883–891[26], both of which were identified by our group, as well as Survivin96–104:M2 [2729], admixed with a pan-HLA-DR tetanus toxoid (TT) peptide (TetA830–845) as an emulsion in a mineral oil base (Montanide), which has been shown in murine models and clinical trials to induce high levels of antigen-specific CTLs,[22, 30] obviating the requirement for harvested dendritic cells as a delivery vehicle. The vaccine was administered adjacent to the immunoadjuvant polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose (poly-ICLC), which has been shown to promote the activity of tumor antigen vaccination in rodent glioma models[30, 31] and to be well tolerated in patients with malignant glioma[22, 3235] and as an adjunct to peptide vaccination in our pilot trial for children with newly diagnosed high-grade and brainstem gliomas[22].

The primary objective of the study was to evaluate the safety and immunological efficacy of vaccination with GAA epitopes in combination with poly-ICLC in pediatric patients with recurrent malignant gliomas. We hypothesized that this regimen would prove safe, and induce anti-glioma immune responses and, potentially, clinical responses.

PATIENT AND METHODS

Patients

Patients between 1 to <22 years of age with recurrent biopsy-confirmed high-grade gliomas (e.g., glioblastoma, anaplastic astrocytoma) that had progressed after prior therapy were eligible for screening. Adequate liver, renal and other organ function, Karnofsky or Lansky performance status > 60, and HLA-A2+ status were required, and patients must have recovered from the effects of prior therapies. At least 3 weeks must have elapsed between the last dose of myelosuppressive therapy (at least 1 week from the last dose of non-myelosuppressive therapy) and initiation of vaccine therapy. The maximal allowable dexamethasone dose during the week before beginning vaccination was 0.1 mg/kg/d (maximum 4 mg/d). The trial was conducted under FDA IND# 13624 and IRB protocol PRO08030085, and was registered with ClinicalTrials.gov (No. NCT01130077). The results in the Strata for newly diagnosed brainstem and high-grade gliomas were previously reported.[22] Signed IRB-approved informed consent was required both for HLA-A2 screening and initiation of therapy.

Study Design

Patients received subcutaneous injections of an emulsion consisting of GAA-derived HLA-A*0201-restricted peptides admixed with a pan-HLA-DR tetanus toxoid (TT) peptide (TetA830–845) in Montanide ISA-51 (Seppic) with adjacent intramuscular injections of the immunoadjuvant poly-ICLC (30 μg/kg, Hiltonol, Oncovir, Inc), a toll-like receptor ligand, every three weeks for a total of 8 vaccines. Aqueous solution (400 μl) containing each HLA-A2-restricted GAA peptide (300 μg/peptide) and the TT peptide (200 μg) was mixed 1:1 with Montanide ISA-51 to form an emulsion (total volume/injection 800 μl). Participants were evaluated for adverse events, regimen-limiting toxicity (RLT), and treatment response by clinic visits, laboratory testing, and MR (magnetic resonance) imaging. Immune response was assessed at 6, 15, and 21 weeks after starting vaccination by ELISPOT assays on peripheral blood mononuclear cells (PBMCs), which were sampled to coincide with MRI scans. Patients demonstrating radiological response (e.g., partial response (PR) or stable disease (SD)) without RLT from the vaccine could continue to receive vaccination at six-week intervals for up to 2 years from the initial vaccine. For such patients, ongoing immunological and MRI evaluation were scheduled to occur at 12-week intervals, in conjunction with every other visit for vaccine administration.

Toxicity Assessment and Stopping Rules

The trial was monitored continuously for treatment-related adverse events using National Cancer Institute Common Toxicity Criteria version 3.0. The following were considered to be RLTs if they were judged to be at least possibly related to treatment: ≥ Grade 2 hypersensitivity or allergic reaction; ≥ Grade 3 non-hematological toxicity; > Grade 3 hematological toxicity that recurred despite 33% poly-ICLC dose reduction or did not resolve to ≤ Grade 1 by the time the next dose was due. The stopping rule for halting accrual for excessive toxicity was an observed rate of RLT of ≥ 33%, provided at least 2 RLTs had been observed.

Peptides

The HLA-A2–restricted peptides that were administered in this study consisted of ALPFGFILV (IL-13Rα2345–353:1A9V)[25]; TLADFDPRV (EphA2883–891)[26]; and LMLGEFLKL (Survivin96–104:M2)[2729]; admixed with a pan-DR helper epitope AQYIKANSKFIGITEL (TetA830–845)[36, 37]. The peptides were synthesized by automated solid-phase peptide synthesis by NeoMPS (PolyPeptide Group, San Diego, CA). Peptides were tested in multiple quality-assurance studies including purity, sterility, identity, potency, pyrogenicity and stability.

ELISPOT assays

Enzyme-linked immunosorbent spot (ELISPOT) assays were performed as described previously[22, 35] on PBMCs obtained and cryopreserved before vaccination (Week 0), and at Weeks 6, 15, 21, and q12 weeks. Briefly, batched cryopreserved Ficoll-isolated PBMC samples were evaluated following in vitro stimulation with IL-13Rα345–353, EphA2883–891, and Survivin96–104 peptides, 10 ng/ml IL-7, and 20 IU/ml IL-2. IFN-γ responses by purified CD8+ and CD4+ T cells were tested against T2 cells pulsed with GAA peptides or PBMCs pulsed with TetA830–845, respectively. A positive ELISPOT response was defined as >2-fold increase in net spot-forming T-cells (after background subtraction) (CD8+ cells for GAAs, CD4+ cells for TetA830–845) over the pre-vaccine level and at least 50 spots/100,000 cells.

Immunohistochemical analysis

In cases with available tissue, expression of the target proteins was determined as previously described [22, 23]. Paraffin-embedded tissue sections were deparaffinized in xylene, and rehydrated in graded concentrations of ethanol. Endogenous peroxidase activity was quenched and antigen retrieval was performed. Non-specific antibody binding was blocked and sections were incubated with monoclonal antibodies against human EphA2 (1:50; Santa Cruz, sc-10746), IL-13Rα2 (1:500; Abcam, ab55275), and Survivin (1:100, sc-10811), followed by EnVision+ System-HRP polymer (Dako). Specimens were graded as positive if they had antigen expression in 20–60% of tumor cells (2) or > 60% of tumor cells (3), versus negative if they had no staining (0) or staining in less than 20% of tumor cells (1), as previously described[22, 23].

Radiological response monitoring: Tumor size was assessed at Weeks 6, 15, 21, and every 12 weeks subsequently by MRI. More frequent scans were obtained if clinically warranted as described below. Response was evaluated by gadolinium-enhanced T1-weighted images, T2-weighted images, or both, based upon the appearance of the tumor on pretreatment MRI. Complete response (CR) was defined as complete disappearance of all enhancing tumor sustained for at least 6 weeks, with no new lesions; stable or improved non-enhancing (T2/FLAIR) lesions; off corticosteroids (or on physiologic replacement doses only); and stable or improved clinically. For non-enhancing tumors, complete disappearance of all T2/FLAIR signal was required. Partial response (PR) was defined as a >50% decrease compared with baseline in the products of the maximal perpendicular diameters of all measurable enhancing lesions (or T2/FLAIR areas) sustained for at least 6 weeks; no new lesions, on stable or declining corticosteroid doses. Minor response was defined as a 25 to < 50% decrease in tumor dimensions compared to baseline, with the above caveats. Progression was defined as >25% increase in sum of the products of perpendicular diameters of the enhancing tumor or T2/FLAIR signal compared with the smallest tumor measurement obtained either at baseline or best response, not explained by pseudoprogression; on stable or increasing doses of corticosteroids; any new lesion. Stable disease (SD) did not qualify for any other above categories.

Management of Immunological Pseudoprogression

In our previous vaccine trials in adults with gliomas[35, 38] and children with newly diagnosed brainstem and high-grade gliomas[22], a subset of participants had pseudoprogression, characterized by transient increase in the size or contrast enhancement of the tumor secondary to intratumoral immune response, followed by tumor stabilization or regression. We therefore incorporated detailed guidelines for pseudoprogression management in this cohort.

If pseudoprogression was suspected following the initiation of vaccination, and the patient was neurologically worse, sufficient to warrant initiation of dexamethasone, subsequent doses of vaccine and poly-ICLC were held. Re-imaging was performed at 4-week intervals until it was determined whether the clinical and imaging changes reflected pseudoprogression or true progression. Once the subject was clinically stable and on <0.1 mg/kg/day decadron for > 1 week, a repeat MRI was performed and the patient could restart treatment with 67% of the poly-ICLC dose (i.e., 20 μg/kg), as long as the MRI changes had improved or resolved.

If the repeat MRI and/or clinical status had not improved, a biopsy or resection would be considered to definitively differentiate between pseudoprogression (i.e., inflammatory/lymphocytic infiltration or necrosis comprising the majority of the specimen) and true tumor progression (where the majority of the resected specimen consisted of tumor). If a biopsy was not deemed to be clinically indicated or safe, the patient was taken off vaccine therapy.

Statistical Methods

This pilot study was designed to enroll children with recurrent high-grade gliomas to assess safety and immunological efficacy. The treatment approach was considered worthy of further investigation if there were at least 5 ELISPOT responses observed among evaluable subjects. In addition, we planned to stop accrual if the rate of RLT was > 33%, and at least 2 RLTs were observed. Patients with disease progression during the first two courses of therapy were replaced for RLT analysis. Survival functions were estimated using the product-limit (Kaplan-Meier) method, and compared by log-rank tests. Greenwood’s method was used to calculate confidence regions for the survival function estimates.

RESULTS

Demographics and Clinical Characteristics

Between November 2009 and September 2015, 69 children with recurrent malignant gliomas were screened for HLA-A2 status, of which 26 (38%) were HLA-A2+. Fourteen of these patients did not enroll, because of progression prior to enrollment (3), ineligible histology (1), metastatic disease (2), poor performance status (1), intracranial hemorrhage (1), travel difficulties (2) or choosing other therapies (4). Twelve children were enrolled (Table 1), 6 with glioblastoma, 5 with anaplastic astrocytoma, and 1 with malignant gliomatosis cerebri. Patients received 2–20 courses of therapy (median 5).

Table 1. Demographics and clinical characteristics of participating patients.

Results of maximal interferon gamma enzyme-linked immunosorbent spot (ELISPOT) reactivity (net spots/105 cells after background subtraction) are shown for each epitope.

Clinical Factors Maximal ELISPOT Responses Immuno-histochemistry Response and Outcome
Patient ID Age at start (years) Gender Tumor Histology Tumor Location Prior Therapy I E S T I E S Best Response Vaccines received PFS OS
1 4.9 F GBM Left Frontal RT w/TMZ NA NA NA NA NA NA NA PD 2 1.0 4.2
2 23.31 M GBM Right Fronto-Parietal RT w/TMZ Bev w/irinotecan 848 796 86 154 NA NA NA SD 9 7.4 21.9
3 11.5 M AA Bithalamic RT w/Bev
Bev w/irinotecan
F F F F 1 2 1 SD 5 4.3 5.8
4 14.7 M GBM Right Occipital Parietal RT
CPM, CDDP, VCR and SCR
602 618 60 1346 1 1 3 PR p study 4 2PTP 12.1
5 19.4 F AA Right Midbrain Thalamus RT
TMZ
8 6 6 10 3 3 NA SD 6 5.5 16.0
6 15.3 F AA Left Frontal RT with SAHA TMZ and Bev Topo/Celebrex/CPM 14 238 12 58 3 3 2 PR 20 39.8+ 39.8+
7 19.1 M AA Left Parieto-Occipital RT w/TMZ and Bev 126 126 62 0 3 1 1 SD 5 3.9 10.8
8 19.9 F GC Basal Ganglia/Chiasm RT w/TMZ and CCNU 46 168 36 6 2 NA NA SD 9 8.0 13.0
9 12.0 F GBM Left Thalamus RT w/cetux and irinotecan TMZ and Bev 12 232 8 44 NA NA NA SD 9 8.6 22.3
10 11.8 F GBM Right Occipital RT
TMZ
2 152 2 4 3 0 0 PD 2 1.8 16.4
11 2.3 M GBM Holocord TMZ 12 290 6 8 3 3 2 PD 2 1.2 4.8
12 10.9 M AA Right Frontal RT
TMZ
50 162 30 2 NA NA NA SD 4 2.8 3.7+

Values ≥50 were considered positive, and reflected at least a two-fold increase compared to prevaccine. Positive responses are shown in bold. NA indicates specimens that were not available because of progression before completion of the second vaccine course. F indicates samples that failed ELISPOT analysis. Immunohistochemistry: pre-vaccine samples with positive expression (e.g., 2 or 3) of GAA targets are indicated in bold; NA indicates patients for whom evaluable tissue was not available. Abbreviations: ID, Identification number; M, male; F, female; GBM – glioblastoma; AA, anaplastic astrocytoma; GC, grade III glioma with gliomatosis cerebri; RT, radiotherapy; TMZ, temozolomide; Bev, Bevacizumab; CPM, cyclophosphamide; CDDP; Cisplatin; VCR, vincristine; SCR, stem cell reconstitution; SAHA, suberoylanilide hydroxamic acid; topo, topotecan; CCNU, lomustine; Cetux, cetuximab; I, IL13Rα2; E, EphA2; S, survivin; Best Response: PR, partial response; SD, stable disease; PD, progressive disease; OS and PFS are expressed in months; PTP, pseudo-tumor progression, also referred to as pseudoprogression.

1

Patient 2 was 20 years of age at the time of screening, but received multiple treatment regimens before beginning vaccine therapy, and was thus >22 at that time.

Summary of Systemic Toxicities

The primary objective of this study was to assess safety, given that this was a component of the first such trial in children. Principal toxicities included Grade 1 and 2 injection site reactions in all 12 patients and flu-like symptoms (such as fatigue, myalgias, fever, chills, headache), which were present in various combinations in virtually all patients, but generally Grade 1 in severity, lasting less than 48 hours after each vaccine and well controlled with acetaminophen or ibuprofen. Grade 1gastrointestinal toxicity was observed in 5 children and Grade 1 anemia and lymphopenia in one each. None of the patients exhibited evidence of autoimmunity.

Pseudoprogression

One child (Patient 4) with a progressive HGG had presumed symptomatic pseudoprogression after the fourth vaccine, with enlargement in tumor size and enhancement that stabilized but did not regress after interruption of protocol therapy. Because repeat resection was not felt to be clinically warranted, vaccine therapy was halted, and the child was withdrawn from the study. The child then received bevacizumab and had a partial response to this agent, ultimately surviving for a year after initial institution of vaccine therapy. This overall course was presumed to be secondary to pseudoprogression.

Induction of Epitope-Specific Immune Responses against GAAs

All but one patient (Patient 1), who had disease progression before completion of the second vaccine cycle, had PBMCs available for immunological analysis. Patient 3’s samples failed analysis. In 9 of 10 evaluable patients (90%), vaccination induced immune reactivity to at least one of the vaccine-targeted GAAs by IFN-γ ELISPOT assays: to IL13Rα2 in 4, EphA2 in 9 and survivin in 3 (Table 1). Three children also had positive responses to the Tet epitope. The time course and magnitude of the ELISPOT responses are summarized in Figure 1A. In some patients, immune responses peaked and then spontaneously dropped prior to or in association with disease progression, possibly reflecting loss of immune response to the vaccine. However, in others responses were maintained over a long interval as illustrated in Figure 1B, in which Patient 6 demonstrated a very prominent response to EphA2 that persisted during the entire 2 year course of vaccination, and also responded to the Tet. Patient 4, who had presumed pseudoprogression, had a particularly strong response to all three vaccine antigens, as well as to the Tet epitope. Because of the small number of patients, it was not possible to demonstrate an association between ELISPOT reactivity and outcome, or between Tet and GAA reactivity.

Figure 1.

Figure 1

A. Time course of glioma-associated antigen (GAA) epitope-specific T-cell responses evaluated by interferon-γ enzyme-linked immunosorbent spot (ELISPOT) analyses in patients that had samples available at week 0 (pre-vaccine) and at the week 6 and 15 time points. Patients lacking pre-vaccine or post-vaccine samples were excluded. Points represent net values after background subtraction. A positive ELISPOT response was defined as >2-fold increase in net spot-forming T-cells (after background subtraction) (CD8+ cells for GAAs, CD4+ cells for TetA830–845) over the pre-vaccine level and at least 50 spots/100,000 cells. IL13Rα and EphA2 are plotted using a logarithmic scale for ELISPOT response to better illustrate the range of values obtained, with a value of 1 used to represent a lack of ELISPOT reactivity (e.g., raw values of 0 or 1).

B. ELISPOT responses in Patient 6, who had a partial response throughout the two-year course of vaccination that has been maintained for more than 39 months. Persistent positive ELISPOT responses to EphA2 were observed throughout the 2-year course of vaccination.

C. Immunohistochemistry results for antigen expression in patients 6 and 7.

Immunohistochemical Analysis

Tissue from prior resections was available for assessing antigen expression in eight children, six of whom were evaluable for all three GAAs. All eight showed immunoreactivity for at least one vaccine antigen: 6 of 8 evaluable had positive staining for IL13Rα2, 4 of 7 for EphA2, and 3 of 6 for survivin. Illustrative results are shown in Figure 1C. Because of the small number of patients, it was not possible to evaluate the association between antigen expression as assessed by immunohistochemistry and ELISPOT reactivity.

Clinical Outcomes

Although the goal of this study was to provide an analysis of safety and tolerability, preliminary efficacy data were obtained. Median progression-free survival (PFS) from the start of vaccination was 4.11 months (95% CI: (2.2,NA)) and median overall survival (OS) was 12.9 months (95% CI: (10.8,NA), Figure 2). Six-month PFS and OS were 33% (95% CI: (0.10,0.65)) and 73% (95% CI: (39%,94%)), respectively. Patient 6 had a partial response that persisted throughout two years of vaccine therapy, and continues at > 39 months (Figure 3). Because of the small numbers of patients in this cohort, an association between ELISPOT response and clinical response was not apparent.

Figure 2.

Figure 2

Kaplan-Meier plots of progression-free (A) and overall (B) survival in 12 recurrent high-grade glioma patients treated on this study.

Figure 3.

Figure 3

MRI results (T1-weighted gadolinium enhanced images) over the course of time in patient 6.

DISCUSSION

This is, to the best of our knowledge, the first clinical report of peptide-based vaccination using a cocktail of GAA-derived epitopes in Montanide, administered in parallel with the immunoadjuvant poly-ICLC in a cohort of children with recurrent childhood high-grade gliomas. Our findings demonstrate reasonable safety of this approach, as well as evidence of immunological responses, and modest clinical activity.

For children with recurrent high-grade gliomas, conventional[4, 5] and molecularly targeted[6, 7] approaches have generally failed to achieve rates of response or 6-month disease stabilization much above 10%. Although recent molecular studies have identified new pharmacological targets for childhood gliomas[3941], these insights have yet to translate into improvements in outcome. Accordingly, novel treatment strategies, such as immunotherapy, warrant consideration.

The GAA epitopes employed in this vaccine were derived from three proteins known to be highly expressed in pediatric gliomas.[23] The immunohistochemistry data obtained in eight patients in the current trial confirmed that these proteins were commonly overexpressed in the treated tumors, consistent with our preliminary data in archival HGG specimens[23]. Likewise, ELISPOT data demonstrated that the majority of vaccinated patients mounted an immune response against at least one of the target antigens, supporting the rationale for incorporating such epitopes in pediatric glioma vaccine trials.

Although our outcome results for patients with recurrent high-grade gliomas suggest that this vaccination approach has at least a modest degree of activity, it is important to emphasize that this was a pilot study focusing on safety, and the requirement for HLA-A2+ status and other restrictive entry criteria may have influenced outcome, considering that only 12 of 69 patients referred for screening received protocol therapy. However, that all patients had progressive disease that had failed to respond to initial and, in some instances, salvage therapy, the 6- month progression-free survival rate of 33% and overall survival of 73% compare favorably to other studies in this population,[5, 42] and the long-term survival in one child that persists beyond 39 months is intriguing. Nonetheless, the results were less encouraging than those observed in our prior studies with recurrent low-grade gliomas[43], which may reflect differences in the immune milieu in these patients.

In that context, the rate of clinical stability or response was notably lower than the frequency of target overexpression and ELISPOT response, and in some patients immunoreactivity waned over time. This phenomenon may reflect a combination of factors, including the presence of immunosuppressive molecules in the tumor itself, the outgrowth of tumor subclones not expressing targeted antigens[44] or antigen processing components, such as MHC molecules[45, 46], or the development of a hostile systemic immune milieu mediated by regulatory T-cell populations[47] or upregulation of immune checkpoint molecules.[48] Elucidating the factors involved will be critical to optimizing the implementation of immunotherapy strategies for these tumors.

The incidence of possible immunological pseudoprogression in this cohort (1 of 12 patients) is lower than we previously observed in a cohort of children with newly diagnosed brainstem gliomas[22], and may reflect distinctive features of tumor response in these two locations. The difficulty we experienced in confirming this diagnosis in the one potentially affected patient highlights the challenges in diagnosing and managing this phenomenon and calls attention to the need for systematically evaluating the utility of advanced imaging techniques, such as a MR spectroscopy and diffusion/perfusion imaging[49, 50], as a way to facilitate this determination.

In summary, this trial demonstrated acceptable safety and promising immunoreactivity in response to peptide vaccination. Although clinical activity was observed, there is clearly room for improvement in our immunotherapy approach in these challenging tumors, perhaps by broadening the panel of antigen epitopes included in the vaccine or by incorporating strategies to prevent immune resistance or escape, such as the use of immune checkpoint inhibitors[48] that can potentiate and maintain immune response to the vaccine.

Acknowledgments

UPCI Clinical Research Services for regulatory management, Andres Salazar, Oncovir, Inc., for provision of poly-ICLC, physicians who referred their patients, and the patients and families who participated in this trial.

FUNDING

This study was supported by National Institutes of Health grants R21CA149872, P01NS40923, and the UPCI Immunologic Monitoring and Cellular Products Laboratory, supported in part by NIH award P30CA47904, and the Pediatric Clinical and Translational Research Center, supported by the NIH through Grant Numbers UL1 RR024153 and UL1TR000005. Support was also provided by grants from the Pediatric Low-Grade Glioma Initiative via the National Brain Tumor Society, the Ellie Kavalieros Fund, the Connor’s Cure Fund, the Ian Yagoda’s Friends Foundation, and the Translational Brain Tumor Fund of the Children’s Hospital of Pittsburgh Foundation.

Footnotes

ETHICAL STATEMENT

Funding sources are as noted above.

Conflicts: Hideho Okada is an inventor in the U.S. Patent Application No. 60,611, 797 (Utility Patent Application) “Identification of An IL-13 Receptor Alpha2 Peptide Analogue Capable of Enhancing Stimulation of Glioma-Specific CTL Response”. An exclusive licensing agreement has been completed on this application between University of Pittsburgh and Stemline, Inc. Due to the potential conflicts of interest, Hideho Okada did not solely interpret any data in the current study. Dr. Regina I. Jakacki is currently employed by Astra Zeneca.

Informed consent: Signed IRB-approved informed consent was required both for HLA-A2 screening and initiation of therapy.

LITERATURE CITED

  • 1.Cohen KJ, Pollack IF, Zhou T, Buxton A, Holmes EJ, Burger PC, Brat DJ, Rosenblum MK, Hamilton RL, Lavey RS, Heideman RL. Temozolomide in the treatment of high-grade gliomas in children: a report from the Children’s Oncology Group. Neuro-oncology. 2011;13:317–323. doi: 10.1093/neuonc/noq191. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.MacDonald TJ, Arenson EB, Ater J, Sposto R, Bevan HE, Bruner J, Deutsch M, Kurczynski E, Luerssen T, McGuire-Cullen P, O’Brien R, Shah N, Steinbok P, Strain J, Thomson J, Holmes E, Vezina G, Yates A, Phillips P, Packer R. Phase II study of high-dose chemotherapy before radiation in children with newly diagnosed high-grade astrocytoma: final analysis of Children’s Cancer Group Study 9933. Cancer. 2005;104:2862–2871. doi: 10.1002/cncr.21593. [DOI] [PubMed] [Google Scholar]
  • 3.Pollack IF, Boyett JM, Yates AJ, Burger PC, Gilles FH, Davis RL, Finlay JL. The influence of central review on outcome associations in childhood malignant gliomas: results from the CCG-945 experience. Neuro-oncology. 2003;5:197–207. doi: 10.1215/S1152-8517-03-00009-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Warren KE, Gururangan S, Geyer JR, McLendon RE, Poussaint TY, Wallace D, Balis FM, Berg SL, Packer RJ, Goldman S, Minturn JE, Pollack IF, Boyett JM, Kun LE. A phase II study of O6-benzylguanine and temozolomide in pediatric patients with recurrent or progressive high-grade gliomas and brainstem gliomas: a Pediatric Brain Tumor Consortium study. J Neurooncol. 2012;106:643–649. doi: 10.1007/s11060-011-0709-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Nicholson HS, Kretschmar CS, Krailo M, Bernstein M, Kadota R, Fort D, Friedman H, Harris MB, Tedeschi-Blok N, Mazewski C, Sato J, Reaman GH. Phase 2 study of temozolomide in children and adolescents with recurrent central nervous system tumors: a report from the Children’s Oncology Group. Cancer. 2007;110:1542–1550. doi: 10.1002/cncr.22961. [DOI] [PubMed] [Google Scholar]
  • 6.Geyer JR, Stewart CF, Kocak M, Broniscer A, Phillips P, Douglas JG, Blaney SM, Packer RJ, Gururangan S, Banerjee A, Kieran MW, Kun LE, Gilbertson RJ, Boyett JM. A phase I and biology study of gefitinib and radiation in children with newly diagnosed brain stem gliomas or supratentorial malignant gliomas. Eur J Cancer. 2010;46:3287–3293. doi: 10.1016/j.ejca.2010.07.005. S0959-8049(10)00662-3 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Fouladi M, Nicholson HS, Zhou T, Laningham F, Helton KJ, Holmes E, Cohen K, Speights RA, Wright J, Pollack IF. A phase II study of the farnesyl transferase inhibitor, tipifarnib, in children with recurrent or progressive high-grade glioma, medulloblastoma/primitive neuroectodermal tumor, or brainstem glioma: a Children’s Oncology Group study. Cancer. 2007;110:2535–2541. doi: 10.1002/cncr.23078. [DOI] [PubMed] [Google Scholar]
  • 8.Okada H, Lieberman FS, Edington HD, Witham TF, Wargo MJ, Cai Q, Elder EH, Whiteside TL, Schold SC, Jr, Pollack IF. Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of recurrent glioblastoma: preliminary observations in a patient with a favorable response to therapy. J Neurooncol. 2003;64:13–20. doi: 10.1007/BF02700016. [DOI] [PubMed] [Google Scholar]
  • 9.Okada H, Lieberman FS, Walter KA, Lunsford LD, Kondziolka DS, Bejjani GK, Hamilton RL, Torres-Trejo A, Kalinski P, Cai Q, Mabold JL, Edington HD, Butterfield LH, Whiteside TL, Potter DM, Schold SC, Jr, Pollack IF. Autologous glioma cell vaccine admixed with interleukin-4 gene transfected fibroblasts in the treatment of patients with malignant gliomas. Journal of translational medicine. 2007;5:67. doi: 10.1186/1479-5876-5-67. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Okada H, Pollack IF. Cytokine gene therapy for malignant glioma. Expert opinion on biological therapy. 2004;4:1609–1620. doi: 10.1517/14712598.4.10.1609. [DOI] [PubMed] [Google Scholar]
  • 11.Okada H, Pollack IF, Lieberman F, Lunsford LD, Kondziolka D, Schiff D, Attanucci J, Edington H, Chambers W, Kalinski P, Kinzler D, Whiteside T, Elder E, Potter D. Gene therapy of malignant gliomas: a pilot study of vaccination with irradiated autologous glioma and dendritic cells admixed with IL-4 transduced fibroblasts to elicit an immune response. Human gene therapy. 2001;12:575–595. doi: 10.1089/104303401300042528. [DOI] [PubMed] [Google Scholar]
  • 12.Okada H, Pollack IF, Lotze MT, Lunsford LD, Kondziolka D, Lieberman F, Schiff D, Attanucci J, Edington H, Chambers W, Robbins P, Baar J, Kinzler D, Whiteside T, Elder E. Gene therapy of malignant gliomas: a phase I study of IL-4-HSV-TK gene-modified autologous tumor to elicit an immune response. Human gene therapy. 2000;11:637–653. doi: 10.1089/10430340050015824. [DOI] [PubMed] [Google Scholar]
  • 13.Kikuchi T, Akasaki Y, Abe T, Fukuda T, Saotome H, Ryan JL, Kufe DW, Ohno T. Vaccination of glioma patients with fusions of dendritic and glioma cells and recombinant human interleukin 12. J Immunother. 2004;27:452–459. doi: 10.1097/00002371-200411000-00005. [DOI] [PubMed] [Google Scholar]
  • 14.Liau LM, Prins RM, Kiertscher SM, Odesa SK, Kremen TJ, Giovannone AJ, Lin JW, Chute DJ, Mischel PS, Cloughesy TF, Roth MD. Dendritic cell vaccination in glioblastoma patients induces systemic and intracranial T-cell responses modulated by the local central nervous system tumor microenvironment. Clin Cancer Res. 2005;11:5515–5525. doi: 10.1158/1078-0432.CCR-05-0464. [DOI] [PubMed] [Google Scholar]
  • 15.Wheeler CJ, Black KL, Liu G, Mazer M, Zhang XX, Pepkowitz S, Goldfinger D, Ng H, Irvin D, Yu JS. Vaccination elicits correlated immune and clinical responses in glioblastoma multiforme patients. Cancer research. 2008;68:5955–5964. doi: 10.1158/0008-5472.CAN-07-5973. [DOI] [PubMed] [Google Scholar]
  • 16.Yamanaka R, Abe T, Yajima N, Tsuchiya N, Homma J, Kobayashi T, Narita M, Takahashi M, Tanaka R. Vaccination of recurrent glioma patients with tumour lysate-pulsed dendritic cells elicits immune responses: results of a clinical phase I/II trial. British journal of cancer. 2003;89:1172–1179. doi: 10.1038/sj.bjc.6601268. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Yamanaka R, Homma J, Yajima N, Tsuchiya N, Sano M, Kobayashi T, Yoshida S, Abe T, Narita M, Takahashi M, Tanaka R. Clinical evaluation of dendritic cell vaccination for patients with recurrent glioma: results of a clinical phase I/II trial. Clin Cancer Res. 2005;11:4160–4167. doi: 10.1158/1078-0432.CCR-05-0120. [DOI] [PubMed] [Google Scholar]
  • 18.Yu JS, Liu G, Ying H, Yong WH, Black KL, Wheeler CJ. Vaccination with tumor lysate-pulsed dendritic cells elicits antigen-specific, cytotoxic T-cells in patients with malignant glioma. Cancer research. 2004;64:4973–4979. doi: 10.1158/0008-5472.CAN-03-3505. 64/14/4973 [pii] [DOI] [PubMed] [Google Scholar]
  • 19.Ardon H, De Vleeschouwer S, Van Calenbergh F, Claes L, Kramm CM, Rutkowski S, Wolff JE, Van Gool SW. Adjuvant dendritic cell-based tumour vaccination for children with malignant brain tumours. Pediatr Blood Cancer. 2010;54:519–525. doi: 10.1002/pbc.22319. [DOI] [PubMed] [Google Scholar]
  • 20.Caruso DA, Orme LM, Neale AM, Radcliff FJ, Amor GM, Maixner W, Downie P, Hassall TE, Tang ML, Ashley DM. Results of a phase 1 study utilizing monocyte-derived dendritic cells pulsed with tumor RNA in children and young adults with brain cancer. Neuro Oncol. 2004;6:236–246. doi: 10.1215/S1152851703000668. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.De Vleeschouwer S, Fieuws S, Rutkowski S, Van Calenbergh F, Van Loon J, Goffin J, Sciot R, Wilms G, Demaerel P, Warmuth-Metz M, Soerensen N, Wolff JE, Wagner S, Kaempgen E, Van Gool SW. Postoperative adjuvant dendritic cell-based immunotherapy in patients with relapsed glioblastoma multiforme. Clin Cancer Res. 2008;14:3098–3104. doi: 10.1158/1078-0432.CCR-07-4875. [DOI] [PubMed] [Google Scholar]
  • 22.Pollack IF, Jakacki RI, Butterfield LH, Hamilton RL, Panigrahy A, Potter DM, Connelly AK, Dibridge SA, Whiteside TL, Okada H. Antigen-specific immune responses and clinical outcome after vaccination with glioma-associated antigen peptides and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in children with newly diagnosed malignant brainstem and nonbrainstem gliomas. J Clin Oncol. 2014;32:2050–2058. doi: 10.1200/JCO.2013.54.0526. JCO.2013.54.0526 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Okada H, Low KL, Kohanbash G, McDonald HA, Hamilton RL, Pollack IF. Expression of glioma-associated antigens in pediatric brain stem and non-brain stem gliomas. J Neurooncol. 2008;88:245–250. doi: 10.1007/s11060-008-9566-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Okano F, Storkus WJ, Chambers WH, Pollack IF, Okada H. Identification of a novel HLA-A*0201-restricted, cytotoxic T lymphocyte epitope in a human glioma-associated antigen, interleukin 13 receptor alpha2 chain. Clin Cancer Res. 2002;8:2851–2855. [PubMed] [Google Scholar]
  • 25.Eguchi J, Hatano M, Nishimura F, Zhu X, Dusak JE, Sato H, Pollack IF, Storkus WJ, Okada H. Identification of interleukin-13 receptor alpha2 peptide analogues capable of inducing improved antiglioma CTL responses. Cancer research. 2006;66:5883–5891. doi: 10.1158/0008-5472.CAN-06-0363. [DOI] [PubMed] [Google Scholar]
  • 26.Hatano M, Eguchi J, Tatsumi T, Kuwashima N, Dusak JE, Kinch MS, Pollack IF, Hamilton RL, Storkus WJ, Okada H. EphA2 as a glioma-associated antigen: a novel target for glioma vaccines. Neoplasia (New York, NY. 2005;7:717–722. doi: 10.1593/neo.05277. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Andersen MH, Pedersen LO, Becker JC, Straten PT. Identification of a cytotoxic T lymphocyte response to the apoptosis inhibitor protein survivin in cancer patients. Cancer research. 2001;61:869–872. [PubMed] [Google Scholar]
  • 28.Andersen MH, Pedersen LO, Capeller B, Brocker EB, Becker JC, thor Straten P. Spontaneous cytotoxic T-cell responses against survivin-derived MHC class I-restricted T-cell epitopes in situ as well as ex vivo in cancer patients. Cancer research. 2001;61:5964–5968. [PubMed] [Google Scholar]
  • 29.Wobser M, Keikavoussi P, Kunzmann V, Weininger M, Andersen MH, Becker JC. Complete remission of liver metastasis of pancreatic cancer under vaccination with a HLA-A2 restricted peptide derived from the universal tumor antigen survivin. Cancer Immunol Immunother. 2005;55:1294–1298. doi: 10.1007/s00262-005-0102-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Zhu X, Nishimura F, Sasaki K, Fujita M, Dusak JE, Eguchi J, Fellows-Mayle W, Storkus WJ, Walker PR, Salazar AM, Okada H. Toll like receptor-3 ligand poly-ICLC promotes the efficacy of peripheral vaccinations with tumor antigen-derived peptide epitopes in murine CNS tumor models. Journal of translational medicine. 2007;5:10. doi: 10.1186/1479-5876-5-10. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Zhu X, Fallert-Junecko B, Fujita M, Ueda R, Kohanbash G, Kastenhuber E, McDonald H, Liu Y, Kalinski P, Reinhart T, Salazar A, Okada H. Poly-ICLC promotes the infiltration of effector T cells into intracranial gliomas via induction of CXCL10 in IFN-α and IFN-γ dependent manners. Cancer Immunology, Immunotherapy. 2010;59:1401–1409. doi: 10.1007/s00262-010-0876-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Butowski N, Chang SM, Junck L, DeAngelis LM, Abrey L, Fink K, Cloughesy T, Lamborn KR, Salazar AM, Prados MD. A phase II clinical trial of poly-ICLC with radiation for adult patients with newly diagnosed supratentorial glioblastoma: a North American Brain Tumor Consortium (NABTC01-05) J Neurooncol. 2009;91:175–182. doi: 10.1007/s11060-008-9693-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Butowski N, Lamborn KR, Lee BL, Prados MD, Cloughesy T, DeAngelis LM, Abrey L, Fink K, Lieberman F, Mehta M, Ian RH, Junck L, Salazar AM, Chang SM. A North American brain tumor consortium phase II study of poly-ICLC for adult patients with recurrent anaplastic gliomas. J Neurooncol. 2009;91:183–189. doi: 10.1007/s11060-008-9705-3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Salazar AM, Levy HB, Ondra S, Kende M, Scherokman B, Brown D, Mena H, Martin N, Schwab K, Donovan D, Dougherty D, Pulliam M, Ippolito M, Graves M, Brown H, Ommaya A. Long-term treatment of malignant gliomas with intramuscularly administered polyinosinic-polycytidylic acid stabilized with polylysine and carboxymethylcellulose: an open pilot study. Neurosurgery. 1996;38:1096–1103. [PubMed] [Google Scholar]
  • 35.Okada H, Kalinski P, Ueda R, Hoji A, Kohanbash G, Donegan TE, Mintz AH, Engh JA, Bartlett DL, Brown CK, Zeh H, Holtzman MP, Reinhart TA, Whiteside TL, Butterfield LH, Hamilton RL, Potter DM, Pollack IF, Salazar AM, Lieberman FS. Induction of CD8+ T-cell responses against novel glioma-associated antigen peptides and clinical activity by vaccinations with {alpha}-type 1 polarized dendritic cells and polyinosinic-polycytidylic acid stabilized by lysine and carboxymethylcellulose in patients with recurrent malignant glioma. J Clin Oncol. 2011;29:330–336. doi: 10.1200/JCO.2010.30.7744. JCO.2010.30.7744 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Slingluff CL, Jr, Yamshchikov G, Neese P, Galavotti H, Eastham S, Engelhard VH, Kittlesen D, Deacon D, Hibbitts S, Grosh WW, Petroni G, Cohen R, Wiernasz C, Patterson JW, Conway BP, Ross WG. Phase I trial of a melanoma vaccine with gp100(280–288) peptide and tetanus helper peptide in adjuvant: immunologic and clinical outcomes. Clin Cancer Res. 2001;7:3012–3024. [PubMed] [Google Scholar]
  • 37.Demotz S, Lanzavecchia A, Eisel U, Niemann H, Widmann C, Corradin G. Delineation of several DR-restricted tetanus toxin T cell epitopes. J Immunol. 1989;142:394–402. [PubMed] [Google Scholar]
  • 38.Okada H, Pollack IF. Do we need novel radiologic response criteria for brain tumor immunotherapy? Expert review of neurotherapeutics. 2011;11:619–622. doi: 10.1586/ern.11.49. [DOI] [PubMed] [Google Scholar]
  • 39.Paugh BS, Qu C, Jones C, Liu Z, Adamowicz-Brice M, Zhang J, Bax DA, Coyle B, Barrow J, Hargrave D, Lowe J, Gajjar A, Zhao W, Broniscer A, Ellison DW, Grundy RG, Baker SJ. Integrated molecular genetic profiling of pediatric high-grade gliomas reveals key differences with the adult disease. J Clin Oncol. 2010;28:3061–3068. doi: 10.1200/JCO.2009.26.7252. JCO.2009.26.7252 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Schwartzentruber J, Korshunov A, Liu XY, Jones DT, Pfaff E, Jacob K, Sturm D, Fontebasso AM, Quang DA, Tonjes M, Hovestadt V, Albrecht S, Kool M, Nantel A, Konermann C, Lindroth A, Jager N, Rausch T, Ryzhova M, Korbel JO, Hielscher T, Hauser P, Garami M, Klekner A, Bognar L, Ebinger M, Schuhmann MU, Scheurlen W, Pekrun A, Fruhwald MC, Roggendorf W, Kramm C, Durken M, Atkinson J, Lepage P, Montpetit A, Zakrzewska M, Zakrzewski K, Liberski PP, Dong Z, Siegel P, Kulozik AE, Zapatka M, Guha A, Malkin D, Felsberg J, Reifenberger G, von Deimling A, Ichimura K, Collins VP, Witt H, Milde T, Witt O, Zhang C, Castelo-Branco P, Lichter P, Faury D, Tabori U, Plass C, Majewski J, Pfister SM, Jabado N. Driver mutations in histone H3.3 and chromatin remodelling genes in paediatric glioblastoma. Nature. 2012;482:226–231. doi: 10.1038/nature10833. nature10833 [pii] [DOI] [PubMed] [Google Scholar]
  • 41.Zarghooni M, Bartels U, Lee E, Buczkowicz P, Morrison A, Huang A, Bouffet E, Hawkins C. Whole-genome profiling of pediatric diffuse intrinsic pontine gliomas highlights platelet-derived growth factor receptor alpha and poly (ADP-ribose) polymerase as potential therapeutic targets. J Clin Oncol. 2010;28:1337–1344. doi: 10.1200/JCO.2009.25.5463. JCO.2009.25.5463 [pii] [DOI] [PubMed] [Google Scholar]
  • 42.Warren KE, Goldman S, Pollack IF, Fangusaro J, Schaiquevich P, Stewart CF, Wallace D, Blaney SM, Packer R, Macdonald T, Jakacki R, Boyett JM, Kun LE. Phase I trial of lenalidomide in pediatric patients with recurrent, refractory, or progressive primary CNS tumors: Pediatric Brain Tumor Consortium study PBTC-018. J Clin Oncol. 2011;29:324–329. doi: 10.1200/JCO.2010.31.3601. JCO.2010.31.3601 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Pollack IF, Jakacki RI, Butterfield LH, Hamilton RL, Panigrahy A, Normolle DP, Connelly AK, Dibridge S, Mason G, Whiteside TL, Okada H. Immune responses and outcome after vaccination with glioma-associated antigen peptides and poly-ICLC in a pilot study for pediatric recurrent low-grade gliomasdagger. Neuro Oncol. 2016 doi: 10.1093/neuonc/now026. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Jager E, Ringhoffer M, Karbach J, Arand M, Oesch F, Knuth A. Inverse relationship of melanocyte differentiation antigen expression in melanoma tissues and CD8+ cytotoxic-T-cell responses: evidence for immunoselection of antigen-loss variants in vivo. Int J Cancer. 1996;66:470–476. doi: 10.1002/(SICI)1097-0215(19960516)66:4&#x0003c;470::AID-IJC10&#x0003e;3.0.CO;2-C. [pii] [DOI] [PubMed] [Google Scholar]
  • 45.van Hall T, Wolpert EZ, van Veelen P, Laban S, van der Veer M, Roseboom M, Bres S, Grufman P, de Ru A, Meiring H, de Jong A, Franken K, Teixeira A, Valentijn R, Drijfhout JW, Koning F, Camps M, Ossendorp F, Karre K, Ljunggren HG, Melief CJ, Offringa R. Selective cytotoxic T-lymphocyte targeting of tumor immune escape variants. Nat Med. 2006;12:417–424. doi: 10.1038/nm1381. nm1381 [pii] [DOI] [PubMed] [Google Scholar]
  • 46.Facoetti A, Nano R, Zelini P, Morbini P, Benericetti E, Ceroni M, Campoli M, Ferrone S. Human leukocyte antigen and antigen processing machinery component defects in astrocytic tumors. Clin Cancer Res. 2005;11:8304–8311. doi: 10.1158/1078-0432.CCR-04-2588. 11/23/8304 [pii] [DOI] [PubMed] [Google Scholar]
  • 47.Gabrilovich DI, Ostrand-Rosenberg S, Bronte V. Coordinated regulation of myeloid cells by tumours. Nat Rev Immunol. 2012;12:253–268. doi: 10.1038/nri3175. nri3175 [pii] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Pardoll DM. The blockade of immune checkpoints in cancer immunotherapy. Nature reviews Cancer. 2012;12:252–264. doi: 10.1038/nrc3239. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Ceschin R, Kurland BF, Abberbock SR, Ellingson BM, Okada H, Jakacki RI, Pollack IF, Panigrahy A. Parametric Response Mapping of Apparent Diffusion Coefficient as an Imaging Biomarker to Distinguish Pseudoprogression from True Tumor Progression in Peptide-Based Vaccine Therapy for Pediatric Diffuse Intrinsic Pontine Glioma. AJNR American journal of neuroradiology. 2015 doi: 10.3174/ajnr.A4428. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Vrabec M, Van Cauter S, Himmelreich U, Van Gool SW, Sunaert S, De Vleeschouwer S, Suput D, Demaerel P. MR perfusion and diffusion imaging in the follow-up of recurrent glioblastoma treated with dendritic cell immunotherapy: a pilot study. Neuroradiology. 2011;53:721–731. doi: 10.1007/s00234-010-0802-6. [DOI] [PubMed] [Google Scholar]

RESOURCES