Abstract
Glioblastoma is an aggressive, often recalcitrant disease. In the majority of cases, prognosis is dismal and current therapies only moderately prolong survival. Immunotherapy is increasingly being recognized as an effective treatment modality. CD70 is a transmembrane protein that shows restricted expression in tissue but has been described in various malignancies. Therapeutic targeting of CD70 has demonstrated antitumor efficacy and is in clinical trials. Here, we sought to characterize CD70 expression in a large cohort of gliomas (n = 205) using tissue microarrays. We identified a subset of tumors (n = 18, 8.8% of high-grade gliomas) exhibiting moderate-to-strong immunoreactivity that enriched for the IDH-wild-type glioblastoma variants gliosarcoma (n = 10) and the newly described epithelioid glioblastoma (n = 4). CD70 expression was associated with prolonged survival in gliosarcoma. Analysis of TCGA datasets showed significantly increased CD70 expression in mesenchymal tumors and prolonged survival in recurrent non-G-CIMP high-expressing tumors. In CD70+ gliomas, there was a significant increase in CD68/CD163/HLA-DR+ tumor-associated macrophages, but not CD27+ TIL. These results confirm prior in vitro studies and demonstrate expression in a clinical cohort. The absence of CD70 expression in the post-treatment setting may portend more clinically aggressive disease in gliosarcoma. However, larger-scale studies will be needed to characterize and validate this relationship.
Keywords: CD70, CD27L, Epithelioid, GBM, Glioblastoma, Gliosarcoma
INTRODUCTION
Novel immunologic approaches have led to significant breakthroughs in cancer therapy in recent years. Notable examples include the cytotoxic T-lymphocyte antigen 4 (CTLA-4) check point inhibitor ipilimumab in unresectable/metastatic melanoma (1), the PD-1 (CD279) inhibitors nivolumab and pembrolizumab in advanced non-small cell lung cancer (NSCLC) and melanoma (2, 3), and chimeric antigen receptor (CAR)-modified T cells targeting CD19 in hematologic malignancies (4, 5). Immunotherapeutic targets under investigation for diffuse gliomas include the mutant epidermal growth factor receptor variant III (EGFRvIII) (6), K27 demethylase in H3.3K27 mutant tumors (7), the R132H mutation in IDH (8), and the PD-1/PD-L1 axis [see (9) for review]. Therapies targeting the co-stimulatory network (eg PD-1/PD-L1, CTLA-4/B7-1) are increasingly being used for the treatment of solid tumors.
CD70 is a type II transmembrane protein and member of the tumor necrosis factor (TNF) family (10, 11). Also known as CD27 ligand (or CD27L), CD70 interacts with its receptor CD27 (constitutively expressed on CD4+ and CD8+ T cells) to regulate cytotoxic T-cell activation, cell proliferation, and survival (12). Expression is normally restricted to activated immune cells and thymic stromal cells (13); however, aberrant expression has been reported in several malignancies, including renal cell carcinoma (14), leukemia/lymphoma (15), NSCLC (16), thymic carcinoma (17), osteosarcoma (18), and melanoma (19). Clinical trials evaluating CD70 as a therapeutic target include the use of antibody–drug conjugates (Clinicaltrials.gov identifier: NCT02216890, NCT01015911), enhanced antibody-dependent cellular cytotoxicity with an anti-CD70 monoclonal antibody (NCT01813539, NCT02759250), and CAR-T cell therapy (NCT02830724).
Expression of CD70 has been demonstrated in glioma cells at both the mRNA and protein level (20, 21). CD70 expression in glioma cells has also been shown to be radio-inducible as a p53-independent response (21). While reports of CD70 protein expression in tissue are limited, immunoreactivity has been shown in anaplastic astrocytoma and glioblastoma (21). An in-depth evaluation and illustration of CD70 expression in a large patient cohort, however, has yet to be reported. Here, we characterize a subset of high-grade gliomas that show strong cell surface expression of CD70, and we describe its association with clinical, histologic, and immunophenotypic features. Given the often marked infiltration of tumor-associated macrophages (TAM) in high-grade gliomas and their association with tumor progression and prognosis (22), we also evaluated the immune microenvironment using markers of macrophage lineage (CD68/CD163/HLA-DR), as well as CD27 expression in T-cells. Lastly, to support our findings, we also surveyed The Cancer Genome Atlas (TCGA) glioma datasets to correlate CD70 mRNA expression levels with overall survival and gene expression-based subtypes. These findings provide evidence from a clinical cohort for the potential therapeutic targeting of CD70 in a subset of high-grade gliomas.
MATERIALS AND METHODS
Case Selection and Tumor Classification
Archival tissues were procured from the National Institutes of Health/National Cancer Institute (NIH/NCI) in Bethesda, Maryland (2000–2016). All patients were deceased at the time of evaluation and were exempt from Institutional Review Board review. At the time of the original diagnosis, cases were classified according to the 2007 WHO Classification of Central Nervous System Tumors (23). Using an integrated diagnostic approach that incorporates histology, immunohistochemistry, and molecular data (where available), cases were re-reviewed and assigned to the newest diagnostic categories outlined in the 2016 WHO classification system (24) (see below for interpretation of immunohistochemistry). Fluorescence in situ hybridization (FISH) for 1p/19q co-deletion was retrospectively included as part of the original case work-up in our lab.
The IDH-wildtype variants gliosarcoma and epithelioid glioblastoma (E-GBM) were diagnosed based on previously reported histologic features (25–7). Briefly, cases of gliosarcoma contained a biphasic population of neoplastic cells consisting of a malignant glial component (GFAP+) and a sarcomatous component composed of overtly neoplastic cells enmeshed in a dense pericellular reticulin meshwork; these 2 areas were often, but not always, regionally distinct. E-GBM contained a monotonous population of round, occasionally discohesive cells with eosinophilic cytoplasm, eccentric nuclei, and prominent nucleoli. No eosinophilic granular bodies or Rosenthal fibers were noted. Focally, areas of spindled cells were occasionally present in an otherwise predominant population of epithelioid cells.
Tissue Microarrays (TMA)
Hematoxylin and eosin (H&E) sections from formalin-fixed paraffin-embedded (FFPE) tissue blocks were evaluated for adequate tumor content. Due to the marked morphologic, immunophenotypic, and molecular heterogeneity in diffuse gliomas, 4-mm tissue cores were used for construction. Tissue cores were extracted from donor paraffin blocks and manually aligned in a melted paraffin bath using a pre-defined grid. Each block contained 28 cores and a 2-mm control tissue marker (liver, kidney, or lymph node) that also served as a spatial reference. H&E-stained sections were then reviewed to confirm representative tumor sampling and accurate core placement.
Immunohistochemistry and Staining Evaluation
Immunohistochemical staining was performed on 4-µm-thick tissue sections with a Ventana BenchMark Ultra (IDH1 R132H, ATRX, p53, H3K27M, EGFR, INI1, GFAP, CD68, CD163, HLA-DR, Ki-67; Ventana Medical Systems, Tucson, AZ), Ventana XT (CD27), or Leica Bond (CD70; Leica Microsystems, Bannockburn, IL) automated immunostainer using manufacturer recommended antigen retrieval. Antibody names, clones, dilutions, source, and expected cellular localization are listed in Table 1. For CD70 staining, a range of dilutions (1:500–1:2000) was tested, with a final dilution of 1:500 used for evaluation. Cases were deemed as having true expression when they exhibited crisp membranous staining or unequivocal strong cytoplasmic staining in tumor cells that was stronger than adjacent non-neoplastic tissue (neuropil, RBCs, vessel media, and adventitia). While a subset of activated lymphocytes express CD70 under normal conditions, absence of staining in adjacent immune cells provided an internal control.
TABLE 1.
Antibodies and Clones Used for Immunohistochemistry
Antibody | Clone | Clonality | Dilution | Source | Localization |
---|---|---|---|---|---|
IDH1 R132H | H09 | Monoclonal | Pre-dilute | Dianova | Cytoplasmic |
ATRX | HPA001906† | Polyclonal | 1:200 | Sigma-Aldrich | Nuclear |
p53 | DO-7 | Monoclonal | 1:1000 | Dako | Nuclear |
H3K27M | ABE419† | Polyclonal | 1:400 | EMD Millipore | Nuclear |
EGFR | 31G7 | Monoclonal | 1:50 | Invitrogen | Membranous/cytoplasmic |
INI1 | MRQ-27 | Monoclonal | Pre-dilute | Roche | Nuclear |
CD70 (CD27L) | MAB2738 | Monoclonal | 1:500 | R&D Systems | Membranous/cytoplasmic* |
CD27 | EPR8569 | Monoclonal | 1:800 | Abcam | Membranous |
CD68 | KP-1 | Monoclonal | Pre-dilute | Roche | Membranous/cytoplasmic |
CD163 | MRQ-26 | Monoclonal | Pre-dilute | Roche | Membranous/cytoplasmic |
HLA-DR | TAL 1B5 | Monoclonal | 1:200 | Dako | Membranous/cytoplasmic |
GFAP | EP672Y | Monoclonal | Pre-dilute | Roche | Cytoplasmic |
CAM5.2 | CAM5.2 | Monoclonal | Pre-dilute | Roche | Cytoplasmic |
See figures for observed staining patterns.
Catalogue number.
HLA-DR, human leukocyte antigen - antigen D related; IDH1, isocitrate dehydrogenase 1; ATRX, alpha thalassaemia-mental retardation, X linked; p53, tumor protein 53; EGFR, epidermal growth factor receptor; GFAP, glial fibrillary acidic protein.
Immunoreactivity for CD70 was assessed using both quantitative and semi-quantitative methods. For semi-quantitative analysis, expression in tumor cells was scored as follows: 0 (negative); 1 (<5% of tumor cells); 2 (5–50% of tumor cells); or 3 (>50% of tumor cells). The cut-off used here (5%) is based on the commonly reported staining threshold used for PD-L1 expression, another co-stimulatory molecule (28). Tumor cells showing weak granular cytoplasmic staining (and a corresponding absence of moderate or strong membranous staining) in any number of tumor cells were given a score of 1. Quantitative methodology is described below. Full tissue sections from positive TMA cases (n = 18) were subsequently stained with anti-CD70 to assess the distribution and heterogeneity of staining. Evaluation of morphologic and immunohistochemical features allowed separation of expression on tumor cells rather than macrophages/microglia to interrogate the association of CD70-positivity and TAM.
IDH mutation status was determined primarily by immunohistochemistry. For designation as IDH-wild-type, a cutoff of 10% was used, where the probability of a variant IDH1 or IDH2 mutation was calculated using a predictive model for IDH mutations in grade II–IV astrocytomas and oligodendrogliomas (29). Cases not meeting the 10% cutoff were designated as not otherwise specified. All gliosarcoma and epithelioid glioblastoma cases were designated as wildtype for IDH; these histologic variants did not show reactivity with anti-IDH R132H. All other surrogate markers were interpreted based on their reported sensitivity and specificity (see Ref. [30] for review). For p53, staining was scored as positive (predictive of p53 mutation) when ≥10% of tumor cells demonstrated moderate or strong nuclear immunoreactivity (31). EGFR was scored as positive when tumor cells showed moderate to strong (2–3+) membranous staining; EGFR immunohistochemistry was mainly used in the context of negative staining (i.e. EGFR amplification is virtually never seen with negative EGFR immunohistochemistry, and has a reported negative predictive value of 0.99–1) (32).
Digital Imaging and Automated Analysis
TMA slides and select case slides from the original paraffin blocks were digitally scanned and converted to whole slide images using the Aperio XT system (Leica Biosystems, Buffalo Grove, IL) at 40× magnification and viewed with Aperio Imagescope software (version 11.2). Tumor areas on TMAs and fresh-cut full sections were digitally annotated with regions of interest for analysis. Staining was quantified using the positive pixel count v9 (ppc; CD70, CD27, and CD163) and nuclear v9 (p53) algorithms and reported as the percentage of positive staining of the tumor area analyzed (irrespective of cellular localization for ppc). Bright-field images were extracted from Imagescope and captured using the Nuance multispectral imaging system (PerkinElmer, Waltham, MA).
BRAF V600E
BRAF V600E testing was performed on FFPE sections of E-GBM cases. DNA was extracted from paraffin-embedded tissue sections using the Qiagen QIAamp DNA FFPE Tissue Kit. The DNA was diluted to an appropriate concentration and subjected to droplet digital PCR for detection of BRAF V600E (c.1799C > A), using a Bio-Rad QX200 Droplet Digital PCR System. In brief, following droplet generation in the QX200 droplet generator, PCR was performed in a 96 well microtiter plate with a single primer set encompassing position c.1799 of the BRAF gene, and amplification products were detected with two allele specific competitive probes, one to wild-type sequence c.1799T (labeled with HEX) and a second to the mutant sequence c.1799A (labeled with FAM) which results in the BRAF p. V600E mutation. Individual droplets were analyzed in the QX200 droplet reader using a 2-color fluorescence detection system and quantitation of the mutant allele fraction was performed with QuantaSoft software. All positive and negative control reactions were adequate. The BRAF V600E (c.1799T > A) ddPCR mutation assay as implemented and validated in our laboratory has a lower limit of detection of 0.1% mutant allele fraction.
TCGA Datasets
Expression levels and clinical data from TCGA (http://cancergenome.nih.gov) and Vital (33) cohort datasets were obtained through the GlioVis data portal for visualization and analysis of brain tumor expression datasets (http://gliovis.bioinfo.cnio.es, accessed May 29, 2017) (34). Using RNAseq and the Affymetrix U133A Array (HG-U133A) data, CD70 mRNA expression levels were acquired from 447 TCGA GBM samples and correlated with overall survival (Affymetrix) and gene expression-based subtype (ie neural, proneural, mesenchymal, and classical) (RNAseq). For generation of survival curves in the recurrent, non-G-CIMP dataset, the median mRNA cutoff (5.32285) was used for defining high and low expression of CD70. Mutation frequency from the TCGA was obtained from cBioPortal (http://www.cbioportal.org, accessed May 29, 2017) (35, 36).
Statistical Analysis
Differences among groups were determined using a Kruskal–Wallis test. Individual 2-group tests were performed using a Wilcoxon rank sum test. In the cases in which several 2-group comparisons were made after using a Kruskal–Wallis test to overall the overall difference among groups, the Hochberg method was used to adjust the p values for multiple comparisons (37). All p values are 2-tailed. Survival analysis was performed using the Kaplan-Meier estimation and log-rank test. For TMA data, survival was calculated (in months) from the date of tumor detection by imaging (overall survival) and the time (months) from the last date of radiotherapy (post-radiation survival) until the date of death. No censored events were included in the TMA analyses; this limited bias is being reported but is not expected to materially impact the interpretation of the findings due to the relatively short survival of patients with glioblastoma following treatment. Analysis of tissue microarray and TCGA data was performed using SAS version 9.2 (SAS Institute, Inc., Cary, NC) and illustrated in Microsoft Excel. A p value <0.05 was considered statistically significant.
RESULTS
Clinicopathologic Features
A total of 214 patients, including 12 paired patient samples (primary and recurrent tissue from the same patient), were included in the initial study cohort. Of these, 9 cases (7 diffuse midline gliomas and 2 IDH-wild-type glioblastomas) showed diminished immunoreactivity with a number of the antibodies tested. In our experience, staining for ATRX (HPA001906) routinely shows decreased immunoreactivity in the presence of tissue artifacts (e.g. prolonged fixation). As a consequence, cases showing loss of normal internal positive control (vessel endothelial cells, non-neoplastic cells such as neurons and glia, etc.) with anti-ATRX were excluded from further analysis. A total of 205 tumor tissues were suitable for evaluation. Patient demographics and clinical characteristics are outlined in Table 2. Classification of tumors according to the 2016 WHO Classification of Central Nervous System Tumors are shown in Table 3. The majority of cases (90.1%) were assigned to the following 2016 WHO diagnostic categories: anaplastic astrocytoma, IDH mutant (n = 23); glioblastoma, IDH mutant (n = 15); glioblastoma, IDH-wild-type (n = 96); glioblastoma, not otherwise specified (n = 29); diffuse midline glioma, H3 K27M mutant (n = 16). A subset of evaluable diffuse midline gliomas were negative for H3K27M by immunohistochemistry (n = 5).
TABLE 2.
Patient Demographics and Clinical Characteristics
CD70 |
||||
---|---|---|---|---|
All | >5% (2–3) | 1–5% (1) | 0 | |
Total cases | 205 | 18 | 48 | 139 |
Unique cases* | 187 | – | – | – |
Age at detection (years)† | ||||
Mean | 44.5 | 47.9 | 48.6 | 42.8 |
Median | 48 | 52 | 50 | 45 |
Range | 4–75 | 16–57 | 16–70 | 4–75 |
Sex† | ||||
Female | 59 | 5 | 12 | 42 |
Male | 126 | 10 | 33 | 83 |
Unknown | 2 | – | – | 2 |
Status‡ | ||||
Primary | 40 | 1 | 13 | 26 |
Recurrence | 163 | 17 | 35 | 113 |
Unknown | 2 | – | – | – |
Laterality (at detection)‡ | ||||
Hemisphere | 179 | 17 | 48 | 116 |
Midline | 21 | 1 | – | 20 |
Both | 2 | – | – | 2 |
Unknown | – | – | – | 1 |
Treatment‡,§ | ||||
Radiotherapy alone | 6 | – | – | 6 |
Chemotherapy alone | 2 | – | – | – |
Chemoradiation only | 21 | 1 | – | 20 |
Resection only | 15 | – | 6 | – |
All | 115 | 13 | 31 | 71 |
None | 32 | 1 | 10 | 21 |
Individual cases minus paired comparisons (ie tumor samples from the same patient taken at different time points).
Unique cases.
Total cases.
Treatment received prior to histologic evaluation.
TABLE 3.
Classification of Tumors According to the 2016 WHO Classification of Central Nervous System Tumors, Revised 4th Edition
2016 WHO classification | CD70 |
|||
---|---|---|---|---|
All | >5% (2–3) | 1–5% (1) | 0 | |
Diffuse astrocytoma, IDH-mut | 3 | – | – | 3 |
Diffuse astrocytoma, NOS | 2 | – | – | 2 |
Anaplastic astrocytoma, IDH-mut | 23 | – | 2 | 21 |
Anaplastic astrocytoma, IDH-wt | 1 | – | – | 1 |
Anaplastic astrocytoma, NOS | 2 | – | – | 2 |
Glioblastoma, IDH-wt | 96 | 16 | 32 | 48 |
Gliosarcoma | 16 | 10 | 5 | 1 |
Epithelioid glioblastoma | 4 | 4 | – | – |
Glioblastoma, IDH-mut | 15 | – | 2 | 13 |
Glioblastoma, NOS* | 29 | – | 10 | 19 |
Diffuse midline glioma, H3 K27M-mutH3 K27M–mut | 16 | 1 | – | 15 |
Diffuse midline glioma, H3 K27M-nonmutH3 K27M–wt | 5 | – | – | 5 |
Oligodendroglioma, NOS | 1 | – | – | 1 |
Anaplastic oligodendroglioma, IDH-mut and 1p/19q-codeleted | 10 | 1 | 1 | 8 |
Anaplastic oligodendroglioma, NOS | 1 | – | – | 1 |
Anaplastic oligoastrocytoma, NOS | 1 | – | 1 | – |
IDH R132H-negative cases without confirmation by PCR, and with a >10% probability of variant IDH mutations, were classified as not otherwise specified (NOS).
A spectrum of staining for CD70 was observed, ranging from focal, weak and cytoplasmic to strong and diffuse cell surface (membranous) expression. Eighteen cases (8.8% of high-grade gliomas) exhibited moderate-to-strong staining (present in >5% of tumor cells and scored as 2 or 3). As a group, these high-expressing cases showed similar histologic and clinical features (see below) and were grouped together for subsequent analysis and illustration. Forty-eight cases (23.4% of all tumors) showed rare weak positive or diffuse faint cytoplasmic staining in tumor cells (scored as 1). Quantitative evaluation of CD70 staining in high-expressing tumors (score 2 to 3) showed a higher median expression of 20.2% (range 4%–68%) compared to a median expression of 0.07% (range 0.0004 to –7.3%) in weak (score 1) or negative (score 0) cases (p < 0.01). Cases with moderate-to-strong (score 2 to 3) CD70 staining exhibited the following characteristics: high-grade histology (WHO grades III and IV, 18/18), history of radiotherapy (13/14), IDH R132H wild-type (17/18), and retained expression of ATRX (14/14) (see Table 4 for complete tumor immunophenotypes).
TABLE 4.
Pathologic Characteristics of CD70+ (2–3) Gliomas
Case | Pathology | IDH1 | ATRX | p53 | H3K27M | EGFR | INI1 | CAM5.2 | Reticulin/ GFAP | BRAF V600E* | CD70 |
|
---|---|---|---|---|---|---|---|---|---|---|---|---|
Semi-quant (score/% of tumor cells) | Quant (ppc)† | |||||||||||
1 | GBIDHWT | − | − | − | − | + | − | + | n/a | n/a | 2/5% | 5.0% |
2 | GS | − | − | + | − | + | − | + | Biphasic | n/a | 2/5% of sarcomatous areas | 5.3% |
3 | GS | − | − | − | − | + | − | + | Biphasic | n/a | 3/50% of sarcomatous areas | 4.0% |
4 | GS | − | − | + | − | − | − | + | Biphasic | n/a | 2/90% of sarcomatous areas | 3.9% |
5 | GS | − | − | − | − | − | − | + | Biphasic | n/a | 3/70% of sarcomatous areas | 16.8% |
6 | DMGH3M | − | − | − | + | − | − | − | n/a | n/a | 3/20% | 17.5% |
7 | E-GBM | − | − | + | − | − | − | − | n/a | Negative | 3/60% of epithelioid tumor cells | 20.6% |
8 | E-GBM | − | − | – | − | − | − | + | n/a | Negative | 3/90% of epithelioid tumor cells | 20.2% |
9 | GS | − | − | + | − | − | − | + | Biphasic | n/a | 3/95% of sarcomatous areas | 28.8% |
10 | GS | − | − | + | − | − | − | − | Biphasic | n/a | 3/90% cytoplasmic in sarcomatous areas | 53.4% |
11 | AOIDHM | + | − | + | − | − | − | − | n/a | n/a | 2/50% | 8.2% |
12 | GS | − | − | – | − | − | − | − | Biphasic | n/a | 2/60% of sarcomatous areas | 4.9% |
13 | GBIDHWT | − | − | − | − | − | − | − | n/a | n/a | 2/10% scattered and peri-necrosis | 21.4% |
14 | E-GBM | − | − | − | − | − | − | + | n/a | Positive | 3/90% membranous and cytoplasmic in epithelioid areas | 67.6% |
15 | GS | − | np | − | np | np | np | np | Biphasic | n/a | 3/80% of sarcomatous areas | 33.7% |
16 | GS | np | np | np | np | np | np | np | Biphasic | n/a | 3/60% of sarcomatous areas | 17.2% |
17 | E-GBM | np | np | np | np | np | np | + | np | Negative | 3/70% of epithelioid tumor cells | 23.0% |
18 | GS | np | np | np | np | np | np | np | Biphasic | n/a | 3/80% of sarcomatous areas | 15.0% |
IDH, ATRX, p53, H3K27M, EGFR, and INI1 were evaluated by IHC; see methods for interpretation of staining.
BRAF V600E mutation tested by ddPCR.
Quantitative analysis of all tumoral areas; this included both sarcomatous and gliomatous components in gliosarcoma cases. This may artificially reduce the ppc since CD70 staining was frequently restricted to sarcomatous areas in gliosarcoma.
Abbreviations: GS, gliosarcoma; E-GBM, epithelioid glioblastoma; DMGH3M, diffuse midline glioma H3 K27M mutant; AOIDHM, anaplastic oligodendroglioma, IDH mutant and 1p19q co-deleted.
n/a, not applicable; np, not performed.
CD70 Is Expressed in the IDH-Wild-type Variants Gliosarcoma and Epithelioid Glioblastoma
A predominant histologic pattern emerged in cases with moderate-to-strong expression, characterized by either focal or diffuse epithelioid cellular morphology (n = 4/4 E-GBM tested; Fig. 1) or sarcomatous transformation (n = 10/16 gliosarcomas tested; Fig. 2). E-GBM is a newly described, but rare, variant of IDH-wildtype glioblastoma with distinct morphologic and molecular features. In our cohort, 4 tumors met histologic criteria for this designation, as it has been recently defined (24) (Fig. 1A–C); BRAF V600E mutation was negative in 3 of these cases. The single case that was V600E positive showed a strong cytoplasmic pattern of staining with focal membranous immunoreactivity, compared to the crisp and diffuse membranous staining observed in the remaining three V600E negative E-GBM cases. Staining for cytokeratins (CAM5.2) was focally present in a majority of CD70-positive tumors, both in gliosarcoma (n = 5) and E-GBM (n = 3) (Fig. 1E). By comparison, only 1 out of 4 CD70-negative gliosarcomas showed immunoreactivity with CAM5.2. Expression of CD70 in gliosarcoma was often restricted to reticulin-rich, GFAP-poor areas (Fig. 2D, I). In our study, all but one gliosarcoma were secondary (ie prior diagnosis of high-grade glioma with subsequent radiation; n = 10/14 with available history). A case of primary gliosarcoma (i.e. arising de novo with no history of high-grade glioma and radiation prior to gliosarcoma diagnosis) was included and showed rare staining for CD70 in <5% of tumor cells. No radiation-induced gliosarcomas (i.e. arising post-radiation without a history of high-grade glioma) were evaluated in this study. No tumors, including those containing an epithelioid morphology, showed evidence of SMARCB1 (hSNF5/INI1) gene alterations (i.e. INI1 expression retained on immunohistochemistry) (Fig. 1F). CD70 expression in an anaplastic oligodendroglioma (IDH-mutant and 1p/19q co-deleted) and a diffuse midline glioma, H3 K27M-mutant tumor was also identified (Fig. 3G–J). Representative images of CD70-negative cases are shown in Supplementary Data Figure S1.
FIGURE 1.
Histologic and immunophenotypic features of CD70+ epithelioid glioblastoma. Three cases of E-GBM showing characteristic round cell morphology and well-delineated cell borders (A–C). E-GBMs demonstrated a characteristic immunophenotype, with patchy expression of GFAP (D) and cytokeratins (E), retained nuclear expression of INI1 (F), and negative IDH R132H (not shown). Strong membranous immunoreactivity for CD70 (1:500, clone MAB2738, R&D Systems) (G–I).
FIGURE 2.
CD70 expression in gliosarcoma. Illustrated are two separate cases of secondary gliosarcoma (A, E) showing the characteristic biphasic features by light microscopy: gliomatous areas highlighted by GFAP (B, F), and a dense peri-cellular reticulin meshwork in the sarcomatous portions of the tumor (C, G). Most gliosarcomas showed expression of CD70 restricted to the mesenchymal/sarcomatous (reticulin-rich/GFAP-poor) portions of the tumor (D, H, I). Half of the gliosarcomas with moderate-to-strong CD70 expression (5/10) also showed focal cytokeratin (CAM5.2) immunoreactivity (not shown).
FIGURE 3.
Staining patterns of CD70 in high-grade gliomas. A prominent Golgi pattern of staining was seen in epithelioid tumor cells (black arrowhead) (A). Isolated dot-like cytoplasmic staining with an absence of membrane staining (B). Cytoplasmic staining in tumor cells was a frequent finding (C). This proved challenging in cases with high-background, but comparison with endothelial expression (black arrowhead) resolved this dilemma. Accordingly, staining of tumor microvasculature (including the entity-defining microvascular proliferation) was a consistent finding (D); only cytoplasmic staining was seen (E) and this was observed in both treatment-naïve and post-radiotherapy tissues. Neurons (black arrowhead) occasionally showed staining of the soma and typically occurred in states of injury (eg perineuronal satellitosis [white arrowhead] and ischemia) (F). CD70 expression in a diffuse midline glioma, H3 K27M mutant (G: anti-CD70; H: anti-H3 K27M-mutant) and an anaplastic oligodendroglioma, IDH-mutant and 1p/19q co-deleted (I: anti-CD70; J: anti-IDH R132H).
Analysis of CD70 expression across the groups using the 2016 WHO classification showed disproportionate, but non-significant, expression in IDH-wild-type glioblastoma (p = 0.11) (Fig. 4A). Stratification by IDH-wild-type variants showed a significantly higher percentage of CD70 expression in gliosarcoma and E-GBM compared to IDH-wild-type glioblastomas with classic histologic features, as well as to the remaining diagnostic entities (p < 0.05; Fig. 4B). In a separate dataset (Vital cohort), mRNA expression was shown to be higher in gliosarcoma (n = 2) compared to glioblastoma samples (n = 26) (Fig. 4C). Additionally, data from the TCGA datasets showed CD70 mRNA expression to be significantly higher in mesenchymal tumors compared to neural, proneural, and classical gene expression-based subtypes (based on subtype signatures by Verhaak et al. (38)) (p < 0.001; Fig. 4D). A significant correlation between CD70 mRNA and protein expression has previously been described (21).
FIGURE 4.
Quantitative analysis of high-grade gliomas with moderate-to-strong (2–3) CD70 expression. Analysis of diagnostic groups showed disproportionate expression in IDH-wildtype glioblastoma (A). Gliosarcoma and E-GBM showed significantly higher expression of CD70 when compared to IDH-wildtype glioblastoma with classic histology (GBIDHWT*), as well as other high-grade diffuse gliomas (*p < 0.05) (B). Evaluation of a small validation dataset (Vital) showed higher CD70 mRNA in gliosarcomas compared to glioblastomas (C). TCGA data showed significantly higher CD70 mRNA expression in mesenchymal tumors compared to other gene expression-based profiles (**p < 0.01) (D).
Expression of CD70 Is Associated With Prior Radiotherapy and Improved Survival in Secondary Gliosarcoma
Of the samples that were obtained after radiation treatment and with available history (n = 14), all but 1 case demonstrated moderate-to-strong CD70 expression (Table 5). Survival analysis showed significantly prolonged post-radiation survival in cases of gliosarcoma with moderate-to-high (2–3) expression of CD70 compared to CD70 low or negative (0–1) gliosarcoma (p < 0.05; Fig. 5A). Overall survival in gliosarcomas based on CD70 expression showed a trend towards increased survival in CD70-positive tumors (groups 2–3 vs groups 0–1, p = 0.062; Fig. 5B). The remaining comparisons showed no significant differences in post-radiation or overall survival. Comparison of paired primary and recurrent tumor samples included CD70-positive (n = 3) and CD70-negative (n = 9) post-radiation samples. In these groups, all treatment naïve tumors (primary diagnosis, n = 12) were negative or showed rare weak cytoplasmic CD70 staining. Hence, 3 paired cases showed new-onset moderate-to-strong expression of CD70 at recurrence after radiotherapy. The time from the last dose of radiation to CD70 staining at recurrence ranged from 6 to 58 months; this was similar to cases without post-radiation CD70 expression (4–50 months). Similar to our findings, analysis of TCGA data showed shorter overall survival in recurrent non-G-CIMP tumors with low CD70 mRNA expression (n = 6, median 17.3) compared to high CD70 mRNA expression (n = 6, median 51.05) (p < 0.01; Fig. 5C).
TABLE 5.
Clinical Characteristics and Treatment History of Cases With Moderate-to-Strong CD70 Expression
At diagnosis |
Interval adjuvant therapy |
At recurrence |
||||||||||
---|---|---|---|---|---|---|---|---|---|---|---|---|
Case | Sex | Age | Location | Pathology | Extent of resection | Chemotherapy | Radiotherapy (modality/total dose) | Age | Location | Pathology | Overall survival† | Survival post-rx‡ |
1 | M | 38 | T | GBM | GTR | GFN | EBR/6000 cGy | 39 | FT | GBIDHWT | 14 | 11 |
2 | M | 41 | T | GBM | P | TMZ,ENZ | GK/1300 cGy | 41 | TP | GS | 8 | 8 |
3 | F | 53 | F | GBM | GTR | TMZ,LAP | EBR/6000 cGy | 54 | F | GS | 28 | 26 |
4 | F | 54 | T | GBM | ST | TMZ | EBR/5940 cGy | 55 | T | GS | 22 | 19 |
5 | M | 52 | P | GBM | GTR | GW,TMZ,CAR,THAL | EBR/6120 cGy | 53 | FT | GS | 12 | 10 |
6 | M | unk | BS | DIPG | unk | unk | unk | unk | unk | DMGH3M | unk | unk |
7 | M | 56 | F | E-GBM* | 14 | 10 | ||||||
8 | M | 57 | P | GBM | GTR | TMZ,BVZ,CRB | EBR/6000 cGy | 57 | T | E-GBM | 21 | 16 |
9 | M | 52 | T | GBM | GTR | TMZ | unknown dose | 54 | T | GS | 30 | 27 |
10 | F | 41 | PO | GBM | GTR | TMZ | EBR/6000 cGy | 44 | FT | GS | 75 | 73 |
11 | M | 42 | F | AO | ST | TMZ,BVZ,CRB,VAN | EBR/4600 cGy | 48 | FP | AO | 70 | 28 |
12 | M | 53 | P | GBM | GTR | TMZ | EBR/6000 cGy | 54 | P | GS | 19 | 17 |
13 | F | 70 | F | GBM | GTR | TMZ | unk | 70 | F | GBIDHWT | 11 | 9 |
14 | M | 16 | M | E-GBM | GTR | TMZ/DAB | EBR/5940 cGy | 17 | T | E-GBM | 16 | 13 |
15 | M | unk | unk | unk | unk | unk | unk | unk | unk | GS | unk | unk |
16 | F | 52 | P | GBM | P | TMZ | EBR/6000 cGy | 53 | P | GS | 27 | 25 |
17 | M | 64 | F | GBM | ST | TMZ,BVZ | EBR/5940 cGy | 70 | F | E-GBM | 63 | 60 |
18 | F | unk | unk | unk | unk | unk | unk | unk | unk | GS | unk | unk |
Primary diagnosis before recurrence/progression.
Overall survival (months) from detection by imaging.
Survival (months) from the last date of radiation (rx) to death.
F, frontal; T, temporal, P, parietal; BS, brainstem; PO, parieto-occipital; TP, temporoparietal; GB, glioblastoma; DIPG, diffuse intrinsic pontine glioma; AO, anaplastic oligodendroglioma; GS, gliosarcoma; E-GBM, epithelioid glioblastoma; DMGH3M, diffuse midline glioma, H3 K27M mutant; ST, subtotal resection; GTR, gross total resection; P, partial resection; GW, Gliadel (carmustine) wafer; GFN, gefitinib; TMZ, temozolomide; ENZ, enzastaurin; LAP, lapatinib; CAR, carmustine; THAL, thalidomide; BVZ, bevacizumab; CRB, carboplatin; VAN, vandetanib; IND, indoximod; DAB, dabrafenib; GK, gamma knife radiation; EBR, external brain radiation.
FIGURE 5.
Kaplan–Meier estimation of survival probability. Gliosarcomas showing moderate-to-strong expression of CD70 (>5% of tumor cells) showed an association with prolonged post-radiation survival compared to gliosarcomas with negative or weak expression, as measured from the last date radiation to death (p < 0.05) (A). Comparison of overall survival in the same groups showed a trend towards significance (p = 0.062) (B). Survival estimation generated from TCGA data in non-G-CIMP tumors showed significantly shorter overall survival in recurrent tumors with low CD70 mRNA expression compared to tumors with high expression (p < 0.01) (C).
CD163+ Tumor-Associated Macrophages Are Significantly Increased in HGG With Moderate-to-Strong Expression of CD70 Without an Accompanying Increase in CD27+ TIL
Expression in all tumors with moderate-to-strong CD70 expression was coupled with a significant increase in CD163+ TAM (p < 0.05; Fig. 6). Staining for CD68 and HLA-DR showed a similar distribution of expression to CD163 (Supplementary Data Fig. S2). Additional comparisons of TAM, including CD70+/− gliosarcomas, were not significant. There was no significant increase in CD27+ TIL in all group comparisons.
FIGURE 6.
Increased TAM in CD70+ gliomas. Post-radiotherapy CD70+ high-grade gliomas showed a significant increase in intratumoral CD163+ macrophages when compared to CD70- tumors (*p < 0.05).
DISCUSSION
Despite therapeutic progress in neuro-oncology, a diagnosis of glioblastoma continues to portend a poor prognosis with a 5-year overall survival recently reported at 5.5% (39). As mentioned previously, the two major classes of immunotherapy, checkpoint inhibitors and adoptive cell therapy, have demonstrated success in treating clinically advanced cancers. Accordingly, immune checkpoint blockade using anti-CTLA4 (tremelimumab, ipilimumab), anti-PD-L1 (durvalumab), and anti-PD1 (nivolumab) therapies are currently being evaluated in clinical trials for glioblastoma (NCT02794883, NCT02311920). Expression of CD70 in glioma cells, and its subsequent interaction with CD27 on B and T cells, has been shown to have immunosuppressive functions on the tumor microenvironment via T-cell apoptosis (21, 40). Anti-tumor efficacy has been demonstrated with targeted therapies against CD70-expressing tumors (41–3). Here, we show that CD70 may be a viable therapeutic target in a subset of gliomas as it is predominantly expressed in the IDH-wild-type glioblastoma variants gliosarcoma and epithelioid glioblastoma. Consistent with in vitro studies, we found that moderate-to-strong expression was largely observed in tumors with prior radiation. In secondary gliosarcomas, we show an association of CD70 expression with improved prognosis following radiation. Expression of CD70 in high-grade gliomas was also accompanied by a significant increase in tumor-associated macrophages.
The CD27–CD70 co-stimulatory axis is highly regulated and serves to stimulate effector and memory T-cells, as well as promote immune cell survival through prevention of apoptosis (44, 45). Expression of CD70 is normally restricted to activated T and B cells, thymic epithelial cells, and dendritic cells in the intestinal mucosa (46). CD70 expression has been demonstrated in 5 of 12 glioblastomas and 3 of 4 anaplastic astrocytomas (21). Reports of CD70 expression in gliomas have shown expression to be preferentially localized within cell processes and in a “stippled” pattern within the cell soma (20); this latter finding is consistent with our own observations with CD70 immunohistochemistry in patient tissue.
Expression of CD70 is inducible by radiation exposure in glioma cells and is thought to alter antitumor responses through proapoptotic effects (21, 47). This mechanism of immune evasion by gliomas was confirmed in a separate study showing attenuation of T-cell death following addition of an anti-CD70 antibody (40). While an association with prior radiation is suggested by our findings in vivo, this may reflect the post-therapeutic nature of cases seen at our institution. It should be noted, however, that 97% (35/36) of treatment-naïve (prior to radiation) high-grade gliomas in our study were negative or only weakly positive for CD70. Furthermore, matched patient tumor samples in our cohort showed expression of CD70 in recurrent post-radiation samples in 3 of 12 cases. Taken together, this suggests that radiation-induced expression of CD70 in high-grade gliomas is more likely to occur in a subset of tumors with specific histopathologic features. Additionally, when accompanied by a sarcomatous phenotype (ie gliosarcoma) at recurrence, moderate-to-strong CD70 expression may signal a better clinical outcome. This will need to be confirmed within a larger cohort of matched pre- and post-radiotherapy samples.
Aberrant expression of CD70 in tumors may be mediated by tumor-specific mechanisms. A recent study identified recurrent mutations, chromosomal deletions, and promoter methylation of the CD70 gene (TNFSF7) as a mechanism of oncogenesis in diffuse large B-cell lymphoma (DLBCL) (48). Promoter methylation has also been implicated in breast cancer progression through silencing of the CD70 gene (49). Additionally, drug-induced overexpression of CD70 has been shown in T-cells through TNFSF7 promoter demethylation (50). Alterations in pVHL, the protein product of the VHL gene, through mutations, epigenetic changes, and hypoxia inducible factor (HIF) deregulation, led to overexpression of CD70 in clear cell RCC (51). To ascertain whether genomic alterations in CD70 occur in high-grade gliomas, we surveyed TCGA datasets (http://www.cbioportal.org) (35, 36). Alterations in CD70 were an infrequent event in high-grade gliomas (0.4%), suggesting other mechanisms for its expression at the protein level. Interaction with the microenvironment, evidenced by the marked increase in macrophages observed in our cohort, suggests possible immune-mediated expression, or may represent a therapy-related response by the tumor cells.
Macrophages and microglia are the predominant immune cell population in gliomas, comprising an estimated 30%–50% of the tumor mass (52, 53). Macrophages are known to promote infiltration and tumor growth through the secretion of cytokines and chemokines such as metalloproteases (MT1-MMP) and colony stimulating factors (see (54) for review). Tumor growth is further potentiated with polarization of activated macrophages to the M2 phenotype. Glioma-associated macrophages generally do not express co-stimulatory molecules (CD70, CD80, and CD86), but express major histocompatibility complex II that is required for T-cell priming in the host anti-tumor response (55). Using markers commonly employed as M2 surrogates, we showed a significant increase in CD163/CD68/HLA-DR+ TAM associated with CD70 expression by tumor cells. The absence of increased CD27+ T-cells in our tumors is consistent with the normal downregulation and shedding of CD27 following interaction with CD70 (56). This interaction also leads to T-cell apoptosis, a mechanism exploited by glioma tumor cells in immune evasion (40). The increased TAM observed here may reflect the overall increase in macrophages seen with escalating grade in diffuse gliomas (57, 58). However, after adjusting for treatment status and grade, we found CD163+ TAM in CD70-expressing high-grade gliomas was significantly greater when compared to CD70-negative tumors. This suggests a possible functional association between TAM and CD70-expressing glioma cells.
Given the relative low frequency of CD70 expression in diffuse gliomas overall, its role as a diagnostic and/or prognostic marker is unclear. Using a 10% threshold of tumor cells with moderate-to-strong staining (a referenced cut-off used in hormone receptor-positive breast cancer), an association with shorter survival and “resistant” disease was noted in CD70+ ovarian carcinomas (59). Shorter overall survival has been reported in DLBCL (both germinal center and activated B-cell like profiles) with high CD70 mRNA expression and in DLBCL (activated B-cell like) with promototer hypomethylation of the CD70 gene (48). In clear cell renal cell carcinoma, shorter survival was seen in tumors with high CD70 expression when associated with increased CD27+ TILs (51). Conversely, no association with CD70 expression and overall survival was seen in head and neck squamous cell carcinoma (60). The association of CD70 expression with survival in post-radiation gliosarcomas raises interesting biologic and therapeutic possibilities. Initial in vitro studies demonstrated CD70-mediated immune cell apoptosis as a mechanism of immune escape in gliomas (21). Subsequent work, however, showed immune stimulatory effects in vivo, and concluded that CD70 expression alone is not sufficient to generate an anti-tumor response (47). Interestingly, a soluble form of CD70 (sCD70) was shown to stimulate a CD8+ T-cell-dependent anti-glioma response in a syngeneic mouse model (61). Taken together, induction of tumor cell killing through targeted therapy (anti-CD70 conjugated antibodies, CAR-T cells) or modulation of the tumor microenvironment may represent potential therapeutic mechanisms in treating CD70-expressing gliomas.
Enrichment of CD70 expression in gliomas with epithelioid or sarcomatous features warrants further investigation in a larger cohort of IDH-wild-type glioblastomas. Furthermore, known molecular alterations in these variants (PTEN and TERT promoter mutations in gliosarcoma, and BRAF V600E mutations in E-GBM) could reveal co-existing immunotherapeutic targets. Lastly, while not all tumors with prior radiotherapy expressed CD70 in our cohort, the marked post-therapeutic expression observed here suggests this could represent a feasible treatment target as part of a multimodal approach.
Supplementary Material
ACKNOWLEDGEMENTS
We would like to thank Dr. Seth Steinberg and Dr. Natasha Pratt for their assistance with the statistical analyses, and Dr. Svetlana Pack and Dr. Zied Abdullaev for their technical assistance with the FISH analyses in the initial work-up of the cases.
REFERENCES
- 1. Schadendorf D, Hodi FS, Robert C, et al. Pooled analysis of long-term survival data from phase II and phase III trials of ipilimumab in unresectable or metastatic melanoma. J Clin Oncol 2015;33:1889–94 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2. Gettinger SN, Horn L, Gandhi L, et al. Overall survival and long-term safety of nivolumab (anti-programmed death 1 antibody, BMS-936558, ONO-4538) in patients with previously treated advanced non-small-cell lung cancer. J Clin Oncol 2015;33:2004–12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Topalian SL, Sznol M, McDermott DF, et al. Survival, durable tumor remission, and long-term safety in patients with advanced melanoma receiving nivolumab. J Clin Oncol 2014;32:1020–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Maude SL, Frey N, Shaw PA, et al. Chimeric antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371:1507–17 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5. Porter DL, Hwang WT, Frey NV, et al. Chimeric antigen receptor T cells persist and induce sustained remissions in relapsed refractory chronic lymphocytic leukemia. Sci Transl Med 2015;7:303ra139 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Swartz AM, Batich KA, Fecci PE, et al. Peptide vaccines for the treatment of glioblastoma. J Neurooncol 2015;123:433–40 [DOI] [PubMed] [Google Scholar]
- 7. Hashizume R, Andor N, Ihara Y, et al. Pharmacologic inhibition of histone demethylation as a therapy for pediatric brainstem glioma. Nat Med 2014;20:1394–6 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Rohle D, Popovici-Muller J, Palaskas N, et al. An inhibitor of mutant IDH1 delays growth and promotes differentiation of glioma cells. Science 2013;340:626–30 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Curry WT, Lim M.. Immunomodulation: Checkpoint blockade etc. Neuro Oncol 2015;17 Suppl 7:vii26–31 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Goodwin RG, Alderson MR, Smith CA, et al. Molecular and biological characterization of a ligand for CD27 defines a new family of cytokines with homology to tumor necrosis factor. Cell 1993;73:447–56 [DOI] [PubMed] [Google Scholar]
- 11. Hintzen RQ, Lens SM, Koopman G, et al. CD70 represents the human ligand for CD27. Int Immunol 1994;6:477–80 [DOI] [PubMed] [Google Scholar]
- 12. Yamada A, Salama AD, Sho M, et al. CD70 signaling is critical for CD28-independent CD8+ T cell-mediated alloimmune responses in vivo. J Immunol 2005;174:1357–64 [DOI] [PubMed] [Google Scholar]
- 13. Tesselaar K, Xiao Y, Arens R, et al. Expression of the murine CD27 ligand CD70 in vitro and in vivo. J Immunol 2003;170:33–40 [DOI] [PubMed] [Google Scholar]
- 14. Diegmann J, Junker K, Gerstmayer B, et al. Identification of CD70 as a diagnostic biomarker for clear cell renal cell carcinoma by gene expression profiling, real-time RT-PCR and immunohistochemistry. Eur J Cancer 2005;41:1794–801 [DOI] [PubMed] [Google Scholar]
- 15. Lens SM, Drillenburg P, den Drijver BF, et al. Aberrant expression and reverse signalling of CD70 on malignant B cells. Br J Haematol 1999;106:491–503 [DOI] [PubMed] [Google Scholar]
- 16. Jacobs J, Zwaenepoel K, Rolfo C, et al. Unlocking the potential of CD70 as a novel immunotherapeutic target for non-small cell lung cancer. Oncotarget 2015;6:13462–75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Hishima T, Fukayama M, Hayashi Y, et al. CD70 expression in thymic carcinoma. Am J Surg Pathol 2000;24:742–6 [DOI] [PubMed] [Google Scholar]
- 18. Pahl JH, Santos SJ, Kuijjer ML, et al. Expression of the immune regulation antigen CD70 in osteosarcoma. Cancer Cell Int 2015;15:31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Pich C, Sarrabayrouse G, Teiti I, et al. Melanoma-expressed CD70 is involved in invasion and metastasis. Br J Cancer 2016;114:63–70 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Held-Feindt J, Mentlein R.. CD70/CD27 ligand, a member of the TNF family, is expressed in human brain tumors. Int J Cancer 2002;98:352–6 [DOI] [PubMed] [Google Scholar]
- 21. Wischhusen J, Jung G, Radovanovic I, et al. Identification of CD70-mediated apoptosis of immune effector cells as a novel immune escape pathway of human glioblastoma. Cancer Res 2002;62:2592–9 [PubMed] [Google Scholar]
- 22. Komohara Y, Ohnishi K, Kuratsu J, et al. Possible involvement of the M2 anti-inflammatory macrophage phenotype in growth of human gliomas. J Pathol 2008;216:15–24 [DOI] [PubMed] [Google Scholar]
- 23. Louis DN, Ohgaki H, Wiestler OD, et al. The 2007 WHO classification of tumours of the central nervous system. Acta Neuropathol 2007;114:97–109 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Louis DN, Perry A, Reifenberger G, et al. The 2016 World Health Organization classification of tumors of the central nervous system: A summary. Acta Neuropathol 2016;131:803–20 [DOI] [PubMed] [Google Scholar]
- 25. Kleinschmidt-DeMasters BK, Aisner DL, Birks DK, et al. Epithelioid GBMs show a high percentage of BRAF V600E mutation. Am J Surg Pathol 2013;37:685–98 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Broniscer A, Tatevossian RG, Sabin ND, et al. Clinical, radiological, histological and molecular characteristics of paediatric epithelioid glioblastoma. Neuropathol Appl Neurobiol 2014;40:327–36 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Miller CR, Perry A.. Glioblastoma. Arch Pathol Lab Med 2007;131:397–406 [DOI] [PubMed] [Google Scholar]
- 28. Patel SP, Kurzrock R.. PD-L1 expression as a predictive biomarker in cancer immunotherapy. Mol Cancer Ther 2015;14:847–56 [DOI] [PubMed] [Google Scholar]
- 29. Chen L, Voronovich Z, Clark K, et al. Predicting the likelihood of an isocitrate dehydrogenase 1 or 2 mutation in diagnoses of infiltrative glioma. Neuro Oncol 2014;16:1478–83 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Tanboon J, Williams EA, Louis DN.. The diagnostic use of immunohistochemical surrogates for signature molecular genetic alterations in gliomas. J Neuropathol Exp Neurol 2016;75:4–18 [DOI] [PubMed] [Google Scholar]
- 31. Takami H, Yoshida A, Fukushima S, et al. Revisiting TP53 mutations and immunohistochemistry—A comparative study in 157 diffuse gliomas. Brain Pathol 2015;25:256–65 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Horbinski C, Hobbs J, Cieply K, et al. EGFR expression stratifies oligodendroglioma behavior. Am J Pathol 2011;179:1638–44 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Vital AL, Tabernero MD, Castrillo A, et al. Gene expression profiles of human glioblastomas are associated with both tumor cytogenetics and histopathology. Neuro Oncol 2010;12:991–1003 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Bowman RL, Wang Q, Carro A, et al. GlioVis data portal for visualization and analysis of brain tumor expression datasets. Neuro Oncol 2017;19:139–41 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Gao J, Aksoy BA, Dogrusoz U, et al. Integrative analysis of complex cancer genomics and clinical profiles using the cBioPortal. Sci Signal 2013;6:pl1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Cerami E, Gao J, Dogrusoz U, et al. The cBio cancer genomics portal: An open platform for exploring multidimensional cancer genomics data. Cancer Discov 2012;2:401–4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37. Guilbaud O. Alternative confidence regions for bonferroni-based closed-testing procedures that are not alpha-exhaustive. Biom J 2009;51:721–35 [DOI] [PubMed] [Google Scholar]
- 38. Verhaak RG, Hoadley KA, Purdom E, et al. Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 2010;17:98–110 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Ostrom QT, Gittleman H, Xu J, et al. CBTRUS statistical report: Primary brain and other central nervous system tumors diagnosed in the United States in 2009-2013. Neuro Oncol 2016;18:v1–v75 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40. Chahlavi A, Rayman P, Richmond AL, et al. Glioblastomas induce T-lymphocyte death by two distinct pathways involving gangliosides and CD70. Cancer Res 2005;65:5428–38 [DOI] [PubMed] [Google Scholar]
- 41. McEarchern JA, Smith LM, McDonagh CF, et al. Preclinical characterization of SGN-70, a humanized antibody directed against CD70. Clin Cancer Res 2008;14:7763–72 [DOI] [PubMed] [Google Scholar]
- 42. Wang QJ, Yu Z, Hanada KI, et al. Preclinical evaluation of chimeric antigen receptors targeting CD70-expressing cancers. Clin Cancer Res 2017;23:2267–76 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Shaffer DR, Savoldo B, Yi Z, et al. T cells redirected against CD70 for the immunotherapy of CD70-positive malignancies. Blood 2011;117:4304–14 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Borst J, Hendriks J, Xiao Y.. CD27 and CD70 in T cell and B cell activation. Curr Opin Immunol 2005;17:275–81 [DOI] [PubMed] [Google Scholar]
- 45. Nolte MA, van Olffen RW, van Gisbergen KP, et al. Timing and tuning of CD27-CD70 interactions: The impact of signal strength in setting the balance between adaptive responses and immunopathology. Immunol Rev 2009;229:216–31 [DOI] [PubMed] [Google Scholar]
- 46. Laouar A, Haridas V, Vargas D, et al. CD70+ antigen-presenting cells control the proliferation and differentiation of T cells in the intestinal mucosa. Nat Immunol 2005;6:698–706 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47. Aulwurm S, Wischhusen J, Friese M, et al. Immune stimulatory effects of CD70 override CD70-mediated immune cell apoptosis in rodent glioma models and confer long-lasting antiglioma immunity in vivo. Int J Cancer 2006;118:1728–35 [DOI] [PubMed] [Google Scholar]
- 48. Bertrand P, Maingonnat C, Penther D, et al. The costimulatory molecule CD70 is regulated by distinct molecular mechanisms and is associated with overall survival in diffuse large B-cell lymphoma. Genes Chromosomes Cancer 2013;52:764–74 [DOI] [PubMed] [Google Scholar]
- 49. Yu SE, Park SH, Jang YK.. Epigenetic silencing of TNFSF7 (CD70) by DNA methylation during progression to breast cancer. Mol Cells 2010;29:217–21 [DOI] [PubMed] [Google Scholar]
- 50. Lu Q, Wu A, Richardson BC.. Demethylation of the same promoter sequence increases CD70 expression in lupus T cells and T cells treated with lupus-inducing drugs. J Immunol 2005;174:6212–9 [DOI] [PubMed] [Google Scholar]
- 51. Ruf M, Mittmann C, Nowicka AM, et al. pVHL/HIF-regulated CD70 expression is associated with infiltration of CD27+ lymphocytes and increased serum levels of soluble CD27 in clear cell renal cell carcinoma. Clin Cancer Res 2015;21:889–98 [DOI] [PubMed] [Google Scholar]
- 52. Charles NA, Holland EC, Gilbertson R, et al. The brain tumor microenvironment. Glia 2011;59:1169–80 [DOI] [PubMed] [Google Scholar]
- 53. Morantz RA, Wood GW, Foster M, et al. Macrophages in experimental and human brain tumors. Part 1: Studies of the macrophage content of experimental rat brain tumors of varying immunogenicity. J Neurosurg 1979;50:298–304 [DOI] [PubMed] [Google Scholar]
- 54. Hambardzumyan D, Gutmann DH, Kettenmann H.. The role of microglia and macrophages in glioma maintenance and progression. Nat Neurosci 2016;19:20–7 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Hussain SF, Yang D, Suki D, et al. The role of human glioma-infiltrating microglia/macrophages in mediating antitumor immune responses. Neuro Oncol 2006;8:261–79 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Hintzen RQ, Lens SM, Beckmann MP, et al. Characterization of the human CD27 ligand, a novel member of the TNF gene family. J Immunol 1994;152:1762–73 [PubMed] [Google Scholar]
- 57. Paulus W, Roggendorf W, Kirchner T.. Ki-M1P as a marker for microglia and brain macrophages in routinely processed human tissues. Acta Neuropathol 1992;84:538–44 [DOI] [PubMed] [Google Scholar]
- 58. Morimura T, Neuchrist C, Kitz K, et al. Monocyte subpopulations in human gliomas: Expression of Fc and complement receptors and correlation with tumor proliferation. Acta Neuropathol 1990;80:287–94 [DOI] [PubMed] [Google Scholar]
- 59. Liu N, Sheng X, Liu Y, et al. Increased CD70 expression is associated with clinical resistance to cisplatin-based chemotherapy and poor survival in advanced ovarian carcinomas. Onco Targets Ther 2013;6:615–9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. De Meulenaere A, Vermassen T, Aspeslagh S, et al. CD70 expression and its correlation with clinicopathological variables in squamous cell carcinoma of the head and neck. Pathobiology 2016;83:327–33 [DOI] [PubMed] [Google Scholar]
- 61. Miller J, Eisele G, Tabatabai G, et al. Soluble CD70: A novel immunotherapeutic agent for experimental glioblastoma. J Neurosurg 2010;113:280–5 [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.