Purpose: Better understanding of the underlying biology of primary central nervous system lymphomas (PCNSL) is critical for the development of early detection strategies, molecular markers, and new therapeutics. This study aimed to define genes associated with survival of patients with PCNSL.

Experimental Design: Expression profiling was conducted on 32 PCNSLs. A gene classifier was developed using the random survival forests model. On the basis of this, prognosis prediction score (PPS) using immunohistochemical analysis is also developed and validated in another data set with 43 PCNSLs.

Results: We identified 23 genes in which expressions were strongly and consistently related to patient survival. A PPS was developed for overall survival (OS) using a univariate Cox model. Survival analyses using the selected 23-gene classifiers revealed a prognostic value for high-dose methotrexate (HD-MTX) and HD-MTX–containing polychemotherapy regimen–treated patients. Patients predicted to have good outcomes by the PPS showed significantly longer survival than those with poor predicted outcomes (P < 0.0001). PPS using immunohistochemical analysis is also significant in test (P = 0.0004) and validation data set (P = 0.0281). The gene-based predictor was an independent prognostic factor in a multivariate model that included clinical risk stratification (P < 0.0001). Among the genes, BRCA1 protein expressions were most strongly associated with patient survival.

Conclusion: We have identified gene expression signatures that can accurately predict survival in patients with PCNSL. These predictive genes should be useful as molecular biomarkers and they could provide novel targets for therapeutic interventions. Clin Cancer Res; 18(20); 5672–81. ©2012 AACR.

Translational Relevance

In this study, we report the development and validation of a risk-score model based on the expression of 23 genes. This 23-gene risk score is highly associated with the outcome of patients with newly diagnosed primary central nervous system lymphoma (PCNSL). These results suggest the importance of this multimarker panel as a stratification factor for the design of future comparative therapeutic trials.

A primary central nervous system lymphoma (PCNSL) is an extranodal form of non-Hodgkin lymphoma arising in the craniospinal axis. For many years, PCNSLs were reported to represent 3% to 5% of all primary central nervous system (CNS) tumors (1). However, PCNSL seems to be increasing in incidence (2–4). The tumor manifestation is often diffuse and multifocal, and most frequently affects the supratentorial brain parenchyma, with periventricular lesions involving the corpus callosum, basal ganglia, or thalamus. The absence of systemic lymphadenopathies and other extracranial localizations of disease should be confirmed. Most PCNSLs belong to the diffuse large B-cell lymphomas (DLBCL) but differ from systemic DLBCLs by their less favorable prognosis.

The systemic use of high-dose methotrexate (HD-MTX)–based chemotherapy with radiotherapy for newly diagnosed PCNSL has improved the median overall survival (OS) from 20 to 36 months (5–8). However, there are still many individual variations within the diagnostic and prognostic categories, resulting in a need for additional biomarkers, partly because of the inability to recognize these patients prospectively. Although, the clinical scoring model using age, Karnofsky performance status (KPS), and lactate dehydrogenase (LDH) level has prognostic value for PCNSL (9–11), it has not been used successfully to stratify patients for therapeutic trials. Molecular markers could improve the outcome prediction, discover potential targets for therapeutic intervention, and elucidate mechanisms that result in resistance to chemotherapy. A comprehensive molecular approach to predict the prognosis is awaited. In the present study, we carried out an expression profiling analysis in patients with PCNSL for the identification of genes that are predictive of OS.

Samples and study population

Patients were diagnosed and treated at Niigata University Hospital (Niigata, Japan), Chiba University Hospital (Chiba, Japan), Yamaguchi University Hospital (Ube, Yamaguchi, Japan), and Toyama Prefectural Central Hospital (Toyama, Japan) between 2000 and 2010. Clinical data were obtained through a registered database and chart review. Inclusion criteria were a histology-proven CNS lymphoma without the evidence of systemic lymphoma, and no evidence of HIV-1 infection, opportunistic infections, or other immunodeficiency. Patients were selected on the basis of the availability of tumor specimens without regard to the clinical outcome. All patients underwent brain imaging with either computed tomography (CT) or magnetic resonance imaging (MRI). After the diagnostic biopsy, detailed history and physical examination, complete blood count, screening blood tests of hepatic and renal function, serum protein electrophoresis, and chest radiographs were obtained. The CT or MRI of the thorax, abdomen, and pelvis were conducted for all patients. Ophthalmologic consultations and slit-lamp examinations were used to rule out ocular involvement. Bone marrow biopsy was not routinely conducted unless CNS involvement was part of a systemic lymphoma. Lumbar puncture for cerebrospinal fluid (CSF) evaluation was routinely conducted. Tissues were snap-frozen in liquid nitrogen within 5 minutes of harvesting, and stored at −80°C thereafter. All specimens were centrally reviewed by a board-certified pathologist by observation of sections of paraffin-embedded tissues that were adjacent or in close proximity to the frozen sample from which the RNA was subsequently extracted. The cut-offs for normal CSF protein and serum LDH levels were 45 mg/dL and 216 IU/L, respectively. Informed consent was obtained from all patients for the use of their samples, in accordance with the guidelines of the respective Ethical Committees on Human Research.

RNA extraction and array hybridization

Approximately, 100 mg of tissue from each tumor was subjected to total RNA extraction using Isogen (Nippon Gene) in accordance with the manufacturer's instructions. These tissues contained more than 95% tumor cells. The quality of the obtained RNA was verified using a Bioanalyzer System (Agilent Technologies) and RNA Pico Chips (Agilent Technologies). Subsequently, 1 μg of RNA was processed for hybridization to a GeneChip Human Genome U133 Plus 2.0 Expression Array (Affymetrix Inc.), which contained approximately 47,000 genes. After hybridization, the chips were processed using a Fluidics Station 450, High-Resolution Microarray Scanner 3000, and GCOS Workstation Version 1.3 (Affymetrix Inc.).

Validation of differential expression by real-time qPCR

Quantitative PCR (qPCR) was conducted using a StepOne Real-Time PCR System (Applied Biosystems) and TaqMan Universal PCR Master Mix (Applied Biosystems) according to the manufacturer's protocol. The Assays-on-Demand probe/primer sets (Applied Biosystems) used were as follows: ATAD1, Hs00907773_g1; BRCA1, Hs01556193_m1; FANCA, Hs01116668_m1; GAPDH, Hs99999905_m1; GGH, Hs00914163_m1; GNASAS, Hs00294858_m1; PGAM1, Hs01652468_g1; PPP3R1, Hs01547793_m1; RBBP8, Hs00161222_m1; ROCK1, Hs01127699_m1; STIL, Hs00161700_m1; TRMT6, Hs00210942_m1; and ZNF681, Hs01862022_s1. Total RNA (1 μg) was reverse-transcribed into cDNA using SuperScript II (Invitrogen), and 1 μL of the resulting cDNA was used for qPCR. Validation was conducted on a subset of tumors that were part of the original tumor data set assessed. Assays were carried out in duplicate. The raw data produced by qPCR referred to the number of cycles required for reactions to reach the exponential phase. Expression of glyceraldehyde-3-phosphate dehydrogenase (GAPDH) was used for normalization of the qPCR data. The mean expression fold change differences between tumor groups were calculated using the 2−ΔΔCT method (12).

Immunohistochemistry

Three antibodies for immunophenotype determination and 5 commercially available antibodies for proteins encoded by genes associated with patient survival were selected for immunohistochemistry (IHC). Sections (5 μm) of the formalin-fixed, paraffin-embedded tissue specimens were evaluated. The primary antibodies recognized BCL6 (DAKO; 1:200 dilution), BRCA1 (Abcam; 1:200 dilution), CD10 (Nichirei; 1:1 dilution), CD79a antibody (DAKO; 1:50 dilution), FANCA (Abcam; 1:3,000 dilution), MUM1 (DAKO; 1:50 dilution), PPP3R1 (Abcam; 1:100 dilution), ROCK1 (Sigma–Aldrich; 1:125 dilution), and RBBP8 (Abnova; 1:200 dilution). Anti-goat or anti-mouse secondary antibodies (Nichirei) were also applied. The staining intensity was classified as none or weakly positive (0 points), moderately positive (1 point), or strongly positive (2 points). The cases in which positive cells were more than 20% of lymphoma cells were determined to be positive. The averages of 3 independent measurements were calculated to the first decimal place. The observers were not aware of the case numbers. For double-immunostaining, after immunostaining of BRCA1 was conducted, sections were washed with glycine buffer (pH 2.2). Then sections were rereacted with anti-CD79a antibody and visualized with HistoGreen (LinarisBiologische).

Bioinformatics analysis

The primary outcome was OS defined as the time from first diagnosis to death or last follow-up. All statistical analyses were conducted using R software (13) and Bioconductor (14). The Affymetrix GeneChip probe-level data were preprocessed using MAS 5.0 (Affymetrix Inc.) for background adjustment and log-transformation (base 2). Each array was normalized by applying a quantile normalization to impose the same empirical distribution of intensities to each array. Genes that passed the filter criteria below were considered for further analysis. To select predictors (genes) for OS, we first set filtered gene expressions and applied the random survival forests-variable hunting (RSF-VH) algorithm (15). Among the parameters in the algorithm, the number of Monte Carlo iterations (nrep) and value controlling the step size used in the forward process (nstep) were set as nrep = 100 and nstep = 5 following Ishwaran and colleagues (15). For other parameters, such as the number of trees and number of variables selected randomly at each node, we used the default settings in the varSelfunction within the RandomSurvivalForest package before the selection. We classified the samples into 2 survival groups by a Ward minimum variance cluster analysis, with inputs of ensemble cumulative hazard functions for each individual for all unique death time points estimated from the fitted random survival forests model to selected genes.

The 2 classified survival groups were used to compute the prognosis prediction score (PPS) from a simple form (linear combination of gene expressions). To do this, we used the principal component analysis and receiver operating characteristic analysis. Briefly, we computed the first principal component of the gene expressions selected by the RSF-VH algorithm as a risk score, and then searched for the optimal cut-off point to predict survival groups with maximum accuracy by the Yoden index (16). The validation for this method was conducted using 10-fold cross-validation. The predictive accuracy of the PPS was assessed by Harrell concordance index.

The Kaplan–Meier method was used to estimate the survival distribution for each group. A log-rank test was used to test the differences between the survival groups. The association of the PPS with OS was evaluated by multivariate analyses with clinical characteristics as other predictors using the Cox proportional hazards regression model. A value of P < 0.05 was considered to indicate statistical significance.

Patient characteristics

The baseline characteristics are shown in Table 1. Expression profiling was conducted on 32 PCNSLs in the test set. The median age of the patients was 64.1 years (range, 44–76 years). Seventeen patients (53%) were males and 15 (47%) were females. The median preoperative KPS was 70. All the patients had histology-proven DLBCL. Twenty-one patients (66%) had a single lesion and 11 (34%) had multiple lesions. Deep structures of the brain, that is, the periventricular lesion, basal ganglia, corpus callosum, brain stem, and/or cerebellum, were involved in 13 patients (41%). Ocular involvement was detected in 4 patients (12.5%), and tumor cell in CSF was positive in 2 patients (6.2%). An elevated LDH serum level was detected in 15 patients (46.8%), and an elevated concentration of CSF protein was detected in 7 of 12 patients (58.3%) assessed. The samples were obtained by stereotactic or open biopsy in 15 patients (46.8%) and surgical resection in 17 patients (53.1%). The treatments were 3 g/m2 per course for 3 or more cycles of HD-MTX in 16 cases (50%) and HD-MTX–containing polychemotherapy (cyclophosphamide, pirarubicin, etoposide, vincristine, procarbazine with or without rituximab; ref. 17) in 16 cases (50%). Chemotherapy alone was used in 8 patients (25%) and chemotherapy followed by radiotherapy in 24 patients (75%). Radiotherapy was administered to the whole brain at 30 Gy and local brain at 20 Gy in 12 patients (37.5%), whole brain at 40 Gy in 9 patients (28.1%) and 20 Gy in 3 patients (9.3%). Relapse after response to the first-line therapy occurred in 19 patients (relapse rate, 59.3%). The second-line treatment was at the physician's choice. The median OS was 1,626 days in all patients. Ten patients (31.2%) remained alive (10 with no evidence of disease) after a median follow-up of 48 months (range, 3.8–135.1 months). The causes of death were lymphoma in 16 patients (72.7%), unrelated causes with no evidence of disease in 3 patients (13.6%), and unknown causes with no evidence of disease in 3 patients (13.6%). The patient characteristics in the validation set are similar to the test set except more patients are involved in deep lesion (Table 1). The patients were monitored for tumor recurrences during the initial and maintenance therapy by MRI or CT.

Table 1.

Patient characteristics

Training set (n = 32)Validation set (n = 43)
CharacteristicN (%)N (%)P
Age, y   0.949 
 Average 64.18 64.34  
 Range 44–76 17–84  
Gender   0.141 
 Male 17 (53) 30 (70)  
 Female 15 (47) 13 (30)  
KPS at diagnosis   0.392 
 Median 70 70   
 70≦ 19 (59) 24 (56)  
 70> 13 (41) 19 (44)  
No. of lesions   0.647 
 Single 21 (66) 26 (60)  
 Multiple 11 (34) 17 (40)  
Deep lesions   <0.05 
 Yes 13 (41) 30 (70)  
 No 19 (59) 13 (30)  
Histology    
 DLBCL 32 (100) 43 (100)  
 Other 0 (0) 0 (0)  
Chemotherapy   0.14 
 HD-MTX 16 (50) 17 (40)  
 Polychemo 16 (50) 23 (53)  
 Other 0 (0) 3 (7)  
Training set (n = 32)Validation set (n = 43)
CharacteristicN (%)N (%)P
Age, y   0.949 
 Average 64.18 64.34  
 Range 44–76 17–84  
Gender   0.141 
 Male 17 (53) 30 (70)  
 Female 15 (47) 13 (30)  
KPS at diagnosis   0.392 
 Median 70 70   
 70≦ 19 (59) 24 (56)  
 70> 13 (41) 19 (44)  
No. of lesions   0.647 
 Single 21 (66) 26 (60)  
 Multiple 11 (34) 17 (40)  
Deep lesions   <0.05 
 Yes 13 (41) 30 (70)  
 No 19 (59) 13 (30)  
Histology    
 DLBCL 32 (100) 43 (100)  
 Other 0 (0) 0 (0)  
Chemotherapy   0.14 
 HD-MTX 16 (50) 17 (40)  
 Polychemo 16 (50) 23 (53)  
 Other 0 (0) 3 (7)  

NOTE: Polychemo, HD-MTX–containing polychemotherapy.

Selection of predictive genes

Microarray data have been deposited in Gene Expression Omnibus (accession number GSE34771). Twenty-three genes were selected as the predictors. Table 2 shows a list of the genes with their obtained variable importance values. Variable importance measures the increase (or decrease) in the prediction error for the random forests model when a variable is randomly “noise up.” That is, if the prediction error of the model became worse when the effect of one variable in the model on the prediction was intentionally destroyed, this means that the variable is important in the model. The scatter plot in Supplementary Fig. S1 shows the relationships between the estimated ensemble mortalities and expressions for 6 selected genes (BRCA1, FANCA, PPP3R1, RBBP8, ROCK1, and ZNF681). Validation of the microarray results was accomplished using qPCR. These 12 genes were also found to be differentially expressed between short-term survivors (survival time, ≤2.5 years; n = 12) and long-term survivors (survival time, ≥4 years; n = 9; Supplementary Table S2).

Table 2.

Identification of survival-related 23 genes

ProbeSymbolDescriptionVI
209092_s_at GLOD4 Glyoxalase domain containing 4 0.0368 
238962_at ZNF681 Zinc finger protein 681 0.0255 
223779_at AFAP1AS AFAP1 antisense RNA (nonprotein coding) 0.0227 
203344_s_at RBBP8 Retinoblastoma binding protein 8 0.0170 
201839_s_at EPCAM Epithelial cell adhesion molecule 0.0170 
236976_at FANCA Fanconi anemia, complementation group A 0.0113 
200886_s_at PGAM1 Phosphoglyceratemutase 1 (brain) 0.0113 
213044_at ROCK1 Rho-associated, coiled-coil containing protein kinase 1 0.0085 
224874_at POLR1D Polymerase (RNA) I polypeptide D, 16 kDa 0.0085 
209146_at SC4MOL Sterol-C4-methyl oxidase-like 0.0085 
239233_at CCDC88A Coiled-coil domain containing 88A 0.0085 
224850_at ATAD1 ATPase family, AAA domain containing 1 0.0057 
236302_at PPM1E Protein phosphatase, Mg2+/Mn2+ dependent, 1E 0.0057 
220176_at NUBPL Nucleotide binding protein-like 0.0057 
204531_s_at BRCA1 Breast cancer 1, early onset 0.0028 
203560_at GGH γ-Glutamyl hydrolase (conjugase, folylpolygammaglutamyl hydrolase) 0.0028 
226103_at NEXN Nexilin (F-actin binding protein) 0.0028 
217398_x_at GAPDH Glyceraldehyde-3-phosphate dehydrogenase 0.0000 
232881_at GNASAS GNAS antisense RNA (nonprotein coding) −0.0028 
223721_s_at DNAJC12 DnaJ (Hsp40) homolog, subfamily C, member 12 −0.0028 
204507_s_at PPP3R1 Protein phosphatase 3, regulatory subunit B, α −0.0057 
205339_at STIL SCL/TAL1 interrupting locus −0.0085 
233970_s_at TRMT6 tRNAmethyltransferase 6 homolog (S. cerevisiae−0.0142 
ProbeSymbolDescriptionVI
209092_s_at GLOD4 Glyoxalase domain containing 4 0.0368 
238962_at ZNF681 Zinc finger protein 681 0.0255 
223779_at AFAP1AS AFAP1 antisense RNA (nonprotein coding) 0.0227 
203344_s_at RBBP8 Retinoblastoma binding protein 8 0.0170 
201839_s_at EPCAM Epithelial cell adhesion molecule 0.0170 
236976_at FANCA Fanconi anemia, complementation group A 0.0113 
200886_s_at PGAM1 Phosphoglyceratemutase 1 (brain) 0.0113 
213044_at ROCK1 Rho-associated, coiled-coil containing protein kinase 1 0.0085 
224874_at POLR1D Polymerase (RNA) I polypeptide D, 16 kDa 0.0085 
209146_at SC4MOL Sterol-C4-methyl oxidase-like 0.0085 
239233_at CCDC88A Coiled-coil domain containing 88A 0.0085 
224850_at ATAD1 ATPase family, AAA domain containing 1 0.0057 
236302_at PPM1E Protein phosphatase, Mg2+/Mn2+ dependent, 1E 0.0057 
220176_at NUBPL Nucleotide binding protein-like 0.0057 
204531_s_at BRCA1 Breast cancer 1, early onset 0.0028 
203560_at GGH γ-Glutamyl hydrolase (conjugase, folylpolygammaglutamyl hydrolase) 0.0028 
226103_at NEXN Nexilin (F-actin binding protein) 0.0028 
217398_x_at GAPDH Glyceraldehyde-3-phosphate dehydrogenase 0.0000 
232881_at GNASAS GNAS antisense RNA (nonprotein coding) −0.0028 
223721_s_at DNAJC12 DnaJ (Hsp40) homolog, subfamily C, member 12 −0.0028 
204507_s_at PPP3R1 Protein phosphatase 3, regulatory subunit B, α −0.0057 
205339_at STIL SCL/TAL1 interrupting locus −0.0085 
233970_s_at TRMT6 tRNAmethyltransferase 6 homolog (S. cerevisiae−0.0142 

Abbreviation: VI, variable importance.

Survival analysis using the selected 23-gene classifiers reveals a prognostic value

Kaplan–Meier curves were drawn for groups classified by clustering analyses based on the gene expressions selected by the significance analysis of microarrays (SAM; ref. 18) with the false discovery rates (Fig. 1A) and by the random survival forests model (Fig. 1B). The corresponding P values by the log-rank test were P = 0.038 for the SAM and P < 0.0001 for the random survival forests model using the 23-gene set. These results show that the random survival forests model is more useful than direct use of the gene expressions.

Figure 1.

Survival analyses using the selected 23-gene classifiers reveal a prognostic value. A, Kaplan–Meier curves comparing groups classified by the clustering analysis based on the gene expressions selected by the SAM. B, comparison of groups classified by the fitted random survival forests model with the 23-gene model. C, Kaplan–Meier curves comparing groups classified by the fitted random survival forests model with the 23-gene model for patients treated with HD-MTX and (D) HD-MTX–containing polychemotherapy. E, Kaplan–Meier curves comparing groups classified by the Z1 PPS with the 23-gene model. F, Kaplan–Meier curves comparing groups classified by the Z2 PPS with age, KPS, PPP3R1, RBBP8, and BRCA1 IHC.

Figure 1.

Survival analyses using the selected 23-gene classifiers reveal a prognostic value. A, Kaplan–Meier curves comparing groups classified by the clustering analysis based on the gene expressions selected by the SAM. B, comparison of groups classified by the fitted random survival forests model with the 23-gene model. C, Kaplan–Meier curves comparing groups classified by the fitted random survival forests model with the 23-gene model for patients treated with HD-MTX and (D) HD-MTX–containing polychemotherapy. E, Kaplan–Meier curves comparing groups classified by the Z1 PPS with the 23-gene model. F, Kaplan–Meier curves comparing groups classified by the Z2 PPS with age, KPS, PPP3R1, RBBP8, and BRCA1 IHC.

Close modal

Survival analysis using the selected 23-gene classifiers reveals a prognostic value independent of HD-MTX–based chemotherapy regimens

The 23-gene profile was tested for the prediction of outcome in the HD-MTX and HD-MTX–containing polychemotherapy groups using Kaplan–Meier curves. The corresponding P values for the log-rank test were P = 0.0001 for the HD-MTX chemotherapy group (Fig. 1C) and P < 0.0001 for the HD-MTX–containing polychemotherapy group (Fig. 1D). These results show that the random survival forests model is useful for predicting survival irrespective of the HD-MTX–based chemotherapy regimens.

Identification of a PPS associated with survival

The PPS was computed from a linear combination of the 23 genes and calculated for each tumor as follows: Z1 = 0.18 × ZNF681 + 0.03 × GNASAS + 0.15 × FANCA + 0.06 × GAPDH + 0.28 × TRMT6 + 0.24 × PGAM1 + 0.28 × PPP3R1 + 0.23 × RBBP8 + 0.27 × ROCK1 + 0.26 × STIL + 0.28 × BRCA1 + 0.26 × ATAD1 + 0.27 × GGH + 0.22 × GLOD4 + 0.15 × EPCAM + 0.14 × AFAP1AS + 0.23 × POLR1D + 0.1 × NEXN + 0.14 × PPM1E + 0.15 × SC4MOL + 0.22 × NUBPL + 0.27 × CCDC88A + 0.04 × DNAJC12.

The Z1-score of the expression value for each individual gene was adapted in this formula. The Z1 scores ranged from −10.0 to 4.43, with a high score associated with a poor outcome. The optimal cut-off was a Z1 score of 1.82. As expected, the predictor performed well in term of the prognosis: the good prognosis group (Z1 ≤ 1.82) had a median survival time of 2,271 days, whereas the poor prognosis group (Z1 > 1.82) had a median survival time of 640 days (P < 0.0001; Fig. 1E). The 10-fold cross-validated c-index was 0.856 [95% confidence interval (CI), 0.824–0.887; P < 0.0001], indicating a significant predictive accuracy.

The potential extension of the microarray-based outcome prediction to the clinical setting was further explored using IHC detection methods. For practical purposes, we tried to incorporate the IHC data and clinical parameters to compute another PPS. The following formula was constructed by the Cox proportional hazards regression model and backward selection:

where KPS was scored as 1 for 70 to 100 and 0 for 10 to 60, and PPP3R1, RBBP8, and BRCA1 were scored as 1 for immunohistochemical scores of 1 or more and 0 for immunohistochemical score of 0.

The Z2 scores ranged from 2.15 to 4.99, with a high score associated with a poor outcome. The optimal cut-off with highest value of the log-rank test was a Z2 score of 3.48. As expected, the predictor performed well in term of the prognosis: the good prognosis group (Z2 ≤ 3.48) had a median survival time of 2,271 days, whereas the poor prognosis group (Z2 > 3.48) had a median survival time of 721 days (P = 0.0004; Fig. 1F). It should be noted that we used different methods to compute the PPS for clinical characteristics owing to poor significance.

The gene expression predictor is the most significant feature

The performance of the gene expression predictor (PPS) was compared with those of traditional individual features. As shown in Table 3, Z1 and Z2 were significantly associated with OS in the univariate analyses. Other prognostic scoring systems, such as IELSG (9) or MSKCC (10) prognostic risk group classification were not significant in our series. Table 4 shows the results for multivariate analyses, in which the clinical characteristics were treated as Z2 as shown in Table 4A or selected by the stepwise procedure as shown in Table 4B. As shown in Table 4, the gene expression predictor Z1 was significantly associated with OS in the multivariate analyses. It should be noted that BRCA1 IHC was associated with progression-free survival (PFS) in the univariate analyses and nearly with OS in the multivariate analyses.

Table 3.

Prognostic value of clinical factors stratified by OS and PFS in patients with PCNSL

VariableMedian OS, wksPMedian PFS, wksP
Age, y  0.581  0.777 
 65≤ 286  178  
 65> 184  73  
Gender  0.324  0.596 
 Male 311  100  
 Female 232  123  
KPS  0.256  0.547 
 70≤ 286  123  
 70> 232  78  
No. of lesions  0.419  0.782 
 Single 286  135  
 Multiple 122  78  
Deep lesions  0.704  0.777 
 Yes 135  68  
 No 286  147  
LDH serum level  0.646  0.199 
 216 ≤ 135  43  
 216> 311  178  
CSF protein level  0.24  0.442 
 Elevated 191  178  
 Normal 324  147  
Operation method  0.26  0.564 
 Biopsy 163  73  
 Removal 286  123  
Chemotherapy  0.733  0.514 
 HD-MTX 136  123  
 Polychemo 298  123  
Immunophenotype (microarray)  0.026  0.098 
 GCB 232  78  
 ABC 79  34  
Immunophenotype (IHC)  0.256  0.578 
 GCB 363  128  
 ABC 135  100  
BRCA1 (IHC)  0.168  0.016 
 Positive 96  35  
 Negative 311  178  
RBBP8 (IHC)  0.232  0.141 
 Positive 135  68  
 Negative 286  147  
ROCK1 (IHC)  0.931  0.94 
 Positive 136  123  
 Negative 271  89  
FANCA (IHC)  0.333  0.197 
 Positive 232  123  
 Negative 122  39  
PPP3R1 (IHC)  0.693  0.401 
 Positive 136  68  
 Negative 311  298  
MSKCC  0.557  0.819 
 Age ≤ 50 495  248  
 Age > 50, 70 ≤ KPS 191  123  
 Age > 50, 70 > KPS 232  47  
IELSG  0.781  0.827 
 0–1 230  111  
 2–3 232  113  
 3–4 223  145  
Z1  <0.0001  <0.0001 
 1.82< 91  34  
 ≤1.82 324  298  
Z2  0.0004  0.0059 
 3.48< 103  41  
 ≤3.48 324  248  
VariableMedian OS, wksPMedian PFS, wksP
Age, y  0.581  0.777 
 65≤ 286  178  
 65> 184  73  
Gender  0.324  0.596 
 Male 311  100  
 Female 232  123  
KPS  0.256  0.547 
 70≤ 286  123  
 70> 232  78  
No. of lesions  0.419  0.782 
 Single 286  135  
 Multiple 122  78  
Deep lesions  0.704  0.777 
 Yes 135  68  
 No 286  147  
LDH serum level  0.646  0.199 
 216 ≤ 135  43  
 216> 311  178  
CSF protein level  0.24  0.442 
 Elevated 191  178  
 Normal 324  147  
Operation method  0.26  0.564 
 Biopsy 163  73  
 Removal 286  123  
Chemotherapy  0.733  0.514 
 HD-MTX 136  123  
 Polychemo 298  123  
Immunophenotype (microarray)  0.026  0.098 
 GCB 232  78  
 ABC 79  34  
Immunophenotype (IHC)  0.256  0.578 
 GCB 363  128  
 ABC 135  100  
BRCA1 (IHC)  0.168  0.016 
 Positive 96  35  
 Negative 311  178  
RBBP8 (IHC)  0.232  0.141 
 Positive 135  68  
 Negative 286  147  
ROCK1 (IHC)  0.931  0.94 
 Positive 136  123  
 Negative 271  89  
FANCA (IHC)  0.333  0.197 
 Positive 232  123  
 Negative 122  39  
PPP3R1 (IHC)  0.693  0.401 
 Positive 136  68  
 Negative 311  298  
MSKCC  0.557  0.819 
 Age ≤ 50 495  248  
 Age > 50, 70 ≤ KPS 191  123  
 Age > 50, 70 > KPS 232  47  
IELSG  0.781  0.827 
 0–1 230  111  
 2–3 232  113  
 3–4 223  145  
Z1  <0.0001  <0.0001 
 1.82< 91  34  
 ≤1.82 324  298  
Z2  0.0004  0.0059 
 3.48< 103  41  
 ≤3.48 324  248  

NOTE: Polychemo, HD-MTX–containing polychemotherapy.

Table 4.

Multivariate analysis: PPS and clinical and therapeutic variables associated with OS

Entire series (n = 32)
VariableSubgroupHR (95% CI)P
(A) 
Z1 Continuous variable 1.73 (1.08–3.52) 0.017 
Z2 Continuous variable 2.49 (0.23–39.4) 0.434 
(B) 
Age Continuous variable 0.99 (0.93–1.05) 0.775 
KPS 70≤, 70> 0.75 (0.28–2.06) 0.573 
BRCA1 (IHC) Positive/negative 2.94 (0.98–8.86) 0.052 
Z1 Continuous variable 1.45 (1.18–1.91) <0.0001 
Entire series (n = 32)
VariableSubgroupHR (95% CI)P
(A) 
Z1 Continuous variable 1.73 (1.08–3.52) 0.017 
Z2 Continuous variable 2.49 (0.23–39.4) 0.434 
(B) 
Age Continuous variable 0.99 (0.93–1.05) 0.775 
KPS 70≤, 70> 0.75 (0.28–2.06) 0.573 
BRCA1 (IHC) Positive/negative 2.94 (0.98–8.86) 0.052 
Z1 Continuous variable 1.45 (1.18–1.91) <0.0001 

Z2 formula was validated in the independent sample set

Because validation of the gene expression signature in another independent set is difficult, Z2 score was validated in the validation set (Table 1). The Z2 scores ranged from 0.97 to 6.36. As expected, there was a significant difference in the OS between the good prognosis group (Z2 ≤ 3.48) and the poor prognosis group (Z2 > 3.48; P = 0.0281; Fig. 2A).

Figure 2.

Z2 PPS and high BRCA1 expression is associated with poor survival. A, Kaplan–Meier curves comparing groups classified by the Z2 PPS in the validation set. B, representative IHC results for BRCA1. The result of double-immunostaining of BRCA1 (brown) and CD79a (B-cell marker, green) showed that BRCA1-positive signals were detected in nucleus of CD79a-positive lymphoma cells, which have enlarged nucleus. C, Kaplan–Meier curves comparing groups classified by BRCA1 IHC for PFS in the test set. D, Kaplan–Meier curves comparing groups classified by BRCA1 IHC for PFS in the validated set.

Figure 2.

Z2 PPS and high BRCA1 expression is associated with poor survival. A, Kaplan–Meier curves comparing groups classified by the Z2 PPS in the validation set. B, representative IHC results for BRCA1. The result of double-immunostaining of BRCA1 (brown) and CD79a (B-cell marker, green) showed that BRCA1-positive signals were detected in nucleus of CD79a-positive lymphoma cells, which have enlarged nucleus. C, Kaplan–Meier curves comparing groups classified by BRCA1 IHC for PFS in the test set. D, Kaplan–Meier curves comparing groups classified by BRCA1 IHC for PFS in the validated set.

Close modal

Classification by cell-of-origin is not associated with survival

We classified our cases in germinal center B-cell–like (GCB) and activated B-cell–like (ABC) subgroups using a gene expression–based method according to Wright and colleagues (19). Ten cases were classified as GCB and 9 were classified as ABC (Supplementary Fig. S3A). There was difference in survival between these 2 groups in univariate analyses (P = 0.026; Supplementary Fig. S3B), and no difference in multivariate analyses (Table 4). We also immunophenotyped the cases in the GCB and ABC subgroups by CD10, BCL-6, and MUM1 IHC according to Camilleri-Broët and colleagues (20). Six cases were classified as GCB and 21 were classified as ABC. There was no difference in survival between these 2 groups (P = 0.256; data not shown). In our cases, classification by cell-of-origin by microarray was not significantly associated with patient survival.

High BRCA1 expression is associated with poor survival

BRCA1 mRNA determined by qPCR was found to be differentially expressed between short-term and long-term survivors (P = 0.027; Supplementary Table S2). Examples of BRCA1 IHC are shown in Fig. 2B. The result of double-immunostaining of BRCA1 and CD79a (B-cell marker) showed that BRCA1-positive signals were detected in nucleus of CD79a-positive lymphoma cells, which had enlarged nucleus. The staining pattern for BRCA1 was predominantly nuclear in 8 cases, cytoplasmic in 7 cases, and both nuclear and cytoplasmic in 4 cases. There was no significant difference in OS between the nuclear and cytoplasmic patterns (data not shown). However, there was a significant difference in the PFS between the BRCA1-positive and BRCA1-negative groups in both datasets (Fig. 2C and D). The PFS was defined as the time from first diagnosis to disease recurrence or death in univariate analyses. Overexpression of BRCA1 mRNA or protein was strongly associated with poor survival in patients with PCNSL. However, FANCA, PPP3R1, ROCK1, and RBBP8 IHC findings were not significantly associated with patient survival (Table 3).

The reason little progress in molecular analyses of PCNSL has been achieved so far is the very tiny sample amounts obtained for genetic analyses. Although, our study is still associated with a small number of patients, it is the largest series to date and the first study using a gene expression prognostic classification context in patients with PCNSL. A better understanding of PCNSL biology is crucial to improve its prognosis. However, only a few studies have been reported on gene expression profiles of PCNSLs. Rubenstein and colleagues (21) compared the gene expression signature of 23 patients with PCNSL with that of 9 patients with nodal large B-cell lymphoma. They showed that individual cases of PCNSL were classified as GCB cell, ABC cell, or type III large B-cell lymphoma based on the cell-of-origin classification described by Alizadeh and colleagues (22). In addition, PCNSLs were distinguished from nodal B-cell lymphoma by high expression of regulators of the unfolded protein response signaling pathway by c-Myc and Pim-1. The IL-4 signaling pathway is associated with tumorigenesis and adverse prognosis in patients with PCNSL (21). Montesinos-Rongen and colleagues (23) reported the gene expression profile of 21 PCNSLs. They showed that PCNSLs resembled late GCB cells in their gene expression pattern, and that PCNSLs were distributed among the spectrum of systemic DLBCLs. Tun and colleagues (24) reported a gene expression comparison between 13 PCNSLs and 30 nonCNS DLBCLs. PCNSL was characterized by significant expression of multiple extracellular matrix- and adhesion-related pathways. Sung and colleagues (25) evaluated 12 patients with PCNSL by comparative genomic hybridization and 7 out of the 12 patients by expression profiling. They selected 8 candidate genes in which expression changes were associated with copy number changes.

Systemic DLBCLs comprise several diseases that differ in responsiveness to chemotherapy (26, 27). The GCB cell–like subgroup expressed genes characteristic of normal GCB cells and were associated with a good outcome, whereas the ABC cell–like subgroup expressed genes characteristic of activated B cells and were associated with a poor outcome. Gene expression analyses of PCNSLs have largely focused on normal lymphocyte development, and the cell-of-origin classification method was not associated with significant survival differences in multivariate analyses. Moreover, prognostic scoring systems, such as IELSG (9) or MSKCC (10) prognostic risk group classification were not significant in our series. Therefore, we developed a novel scoring system based on molecular markers.

We assessed the relationships between gene expressions and survival time using the random survival forests model, and its performance provided a better classification compared with the SAM and gene expression subgroups. As discussed in a study by Cordell (28), the functional form should contain gene-by-gene interaction terms. The random forests method is classified into a tree-based method, which has an advantage in detecting interactions. It has been developed for application to data with several variables (genes) much larger than the number of patients. In this regard, a framework of random forests that overcomes this problem would be necessary in the analysis. Genes were selected by applying the RSF-VH algorithm. The advantage of this method is that no screening of the genes is necessary. There are many studies on microarray data using univariate analyses for screening, in which potential genes interacting with other genes may be dropped from the analyses. In this regard, the RSF-VH algorithm would be more desirable.

Among the selected genes, PPP3R1 is a calmodulin-regulated protein phosphatase, which plays an important role in signal transduction (29), although there are no reports about its role in cancer development. BRCA1 seems to promote cell survival after DNA damage by preventing apoptosis and participating in repair pathways (30). In addition, a role of BRCA1 in the cellular response to chemotherapy has been discussed (31, 32). The expression levels of BRCA1 mRNA or protein predicted survival after chemotherapy for patients with sporadic ovarian (33), breast (34, 35), prostate (36), and non–small cell lung cancer (37–40). Low levels of BRCA1 expression resulted in increased sensitivity to platinum therapy and decreased sensitivity to taxane therapy (33, 35, 37–40). Silencing of the BRCA1 gene by promoter hypermethylation was reported in sporadic breast and ovarian tumors, especially in the presence of loss of heterogeneity (41). We have provided evidence that BRCA1 expression may represent a predictive biomarker of survival in patients with PCNSL. We are trying to further investigate the associations between BRCA1 expression and the chemotherapeutic response by MTX. Furthermore, as BRCA1 shows promise as a prognostic and predictive marker in PCNSL, patients identified as being high expressors could be treated with agents that downregulate BRCA1, thereby sensitizing them to standard therapies. RBBP8 (also known as CtIP) is a BRCA1-interacting protein (42) and implicated the functional involvement in the development of tamoxifen resistance for breast cancer (43). Furthermore, work, both in vitro studies and clinical trials, is needed to assess the correlations between BRCA1-RBBP8 complex expression levels and responses to potential-targeted therapies.

Because these are retrospective analyses, there are limitations and other limitations inherent in a retrospective design. So, these results should be investigated further in the future. Our PPS may help to identify the patients with PCNSL who are unlikely to be cured by standard therapy. Our PPS involves a small number of genes, and thus quantitative reverse transcriptase PCR assays or customized DNA microarrays could be developed for clinical applications. Much more aggressive therapies, such as high-dose chemotherapy with stem cell implantation (44) or molecular-targeted therapies that specifically target disabled pathways, might be tailored in those patients with a poor prognosis. In this regard, the expression profiles might not only predict the likelihood of short-term survival, but also yield clues on individual genes involved in tumor development, progression, and response to therapy. Moreover, the ability to distinguish PCNSLs will enable appropriate therapies to be tailored to specific tumor subtypes. Class prediction models based on defined molecular profiles allow classification of PCNSLs in a manner that will be better correlated with clinical outcomes. Therefore, identification of these molecular subclasses of PCNSLs could greatly facilitate prognosis prediction and our ability to develop effective treatment protocols. In conclusion, our profiling results will help to construct a new classification scheme that better assesses these clinical malignancies.

No potential conflicts of interest were disclosed.

Conception and design: R. Yamanaka

Development of methodology: A. Kawaguchi, Y. Komohara, N. Tsuchiya, R. Yamanaka

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): Y. Iwadate, M. Sano, K. Kajiwara, N. Yajima, N. Tsuchiya, J. Homma, H. Aoki, T. Kobayashi, Y. Sakai, H. Hondoh, Y. Fujii, R. Yamanaka

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): A. Kawaguchi, Y. Iwadate, T. Kakuma, R. Yamanaka

Writing, review, and/or revision of the manuscript: A. Kawaguchi, R. Yamanaka

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Kawaguchi, Y. Sakai, Y. Fujii, R. Yamanaka

Study supervision: R. Yamanaka

Pathological diagnosis, analyzation, and interpretation of the immunohistochemical data: Y. Komohara

Carrying out experiments and analyzing data: Y. Sakai

The authors thank Akiyoshi Kakita of Resource Branch for Brain Disease Research, Brain Research Institute, Niigata University for preparing specimens.

This work was supported in part by JSPS KAKENHI grant number 21700312 to A. Kawaguchi and 20390392 to R. Yamanaka, and by the Collaborative Research Project grant number 2010–2022 to R. Yamanaka of the Brain Research Institute, Niigata University.

The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact.

1.
Panageas
KS
,
Elkin
EB
,
DeAngelis
LM
,
Ben-Porat
L
,
Abrey
LE
. 
Trends in survival from primary central nervous system lymphoma, 1975–1999: a population-based analysis
.
Cancer
2005
;
104
:
2466
72
.
2.
Olson
JE
,
Janney
CA
,
Rao
RD
,
Cerhan
JR
,
Kurtin
PJ
,
Schiff
D
, et al
The continuing increase in the incidence of primary central nervous system non-Hodgkin lymphoma: a surveillance, epidemiology, and end results analysis
.
Cancer
2002
;
95
:
1504
10
.
3.
Haldorsen
IS
,
Krossnes
BK
,
Aarseth
JH
,
Scheie
D
,
Johannesen
TB
,
Mella
O
, et al
Increasing incidence and continued dismal outcome of primary central nervous system lymphoma in Norway 1989–2003: time trends in a 15-year national survey
.
Cancer
2007
;
110
:
1803
14
.
4.
Makino
K
,
Nakamura
H
,
Kino
T
,
Takeshima
H
,
Kuratsu
J
. 
Rising incidence of primary central nervous system lymphoma in Kumamoto, Japan
.
Surg Neurol
2006
;
66
:
503
6
.
5.
Gavrilovic
IT
,
Hormigo
A
,
Yahalom
J
,
DeAngelis
LM
,
Abrey
LE
. 
Long-term follow-up of high-dose methotrexate-based therapy with and without whole brain irradiation for newly diagnosed primary CNS lymphoma
.
J Clin Oncol
2006
;
24
:
4570
4
.
6.
Ferreri
AJ
,
Reni
M
,
Foppoli
M
,
Martelli
M
,
Pangalis
GA
,
Frezzato
M
, et al
High-dose cytarabine plus high-dose methotrexate versus high-dose methotrexate alone in patients with primary CNS lymphoma: a randomised phase 2 trial
.
Lancet
2009
;
374
:
1512
20
.
7.
Chamberlain
MC
,
Johnston
SK
. 
High-dose methotrexate and rituximab with deferred radiotherapy for newly diagnosed primary B-cell CNS lymphoma
.
Neuro Oncol
2010
;
12
:
736
44
.
8.
Thiel
E
,
Korfel
A
,
Martus
P
,
Kanz
L
,
Griesinger
F
,
Rauch
M
, et al
High-dose methotrexate with or without whole brain radiotherapy for primary CNS lymphoma (G-PCNSL-SG-1): a phase 3, randomised, non-inferiority trial
.
Lancet Oncol
2010
;
11
:
1036
47
.
9.
Ferreri
AJ
,
Blay
JY
,
Reni
M
,
Pasini
F
,
Spina
M
,
Ambrosetti
A
, et al
Prognostic scoring system for primary CNS lymphomas: the International Extranodal Lymphoma Study Group experience
.
J Clin Oncol
2003
;
21
:
266
72
.
10.
Abrey
LE
,
Ben-Porat
L
,
Panageas
KS
,
Yahalom
J
,
Berkey
B
,
Curran
W
, et al
Primary central nervous system lymphoma: the Memorial Sloan-Kettering Cancer Center prognostic model
.
J Clin Oncol
2006
;
24
:
5711
5
.
11.
Shenkier
TN
,
Voss
N
,
Chhanabhai
M
,
Fairey
R
,
Gascoyne
RD
,
Hoskins
P
, et al
The treatment of primary central nervous system lymphoma in 122 immunocompetent patients: a population-based study of successively treated cohorts from the British Colombia Cancer Agency
.
Cancer
2005
;
103
:
1008
17
.
12.
Livak
KJ
,
Schmittgen
TD
. 
Analysis of relative gene expression data using real-time quantitative PCR and the 2(−Delta Delta C(T)) method
.
Methods
2001
;
25
:
402
8
.
13.
R Development Core Team
. 
R. A language and environment for statistical computing
.
Vienna, Austria
:
R Foundation for Statistical Computing
; 
2009
.
Available from
: www.R-project.org.
14.
Gentleman
R
,
Carey
V
,
Bates
D
,
Bolstad
B
,
Dettling
M
,
Dudoit
S
, et al
Bioconductor: open software development for computational biology and bioinformatics
.
Genome Biol
2004
;
5
:
R80
.
15.
Ishwaran
H
,
Kogalur
UB
,
Gorodeski
EZ
,
Minn
AJ
,
Lauer
MS
. 
High-dimensional variable selection for survival data
.
J Am Stat Assoc
2010
;
105
:
205
17
.
16.
Youden
WJ
. 
Index for rating diagnostic tests
.
Cancer
1950
;
3
:
32
5
.
17.
Yamanaka
R
,
Shinbo
Y
,
Sano
M
,
Homma
J
,
Tsuchiya
N
,
Yajima
N
, et al
Salvage therapy and late neurotoxicity in patients with recurrent primary CNS lymphoma treated with a modified ProMACE-MOPP hybrid regimen
.
Leuk Lymphoma
2007
;
48
:
1119
26
.
18.
Tusher
VG
,
Tibshirani
R
,
Chu
G
. 
Significance analysis of microarrays applied to the ionizing radiation response
.
Proc Natl Acad Sci U S A
2001
;
98
:
5116
21
.
19.
Wright
G
,
Tan
B
,
Rosenwald
A
,
Hurt
EH
,
Wiestner
A
,
Staudt
LM
. 
A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma
.
Proc Natl Acad Sci U S A
2003
;
100
:
9991
6
.
20.
Camilleri-Broët
S
,
Crinière
E
,
Broët
P
,
Delwail
V
,
Mokhtari
K
,
Moreau
A
, et al
A uniform activated B-cell-like immunophenotype might explain the poor prognosis of primary central nervous system lymphomas: analysis of 83 cases
.
Blood
2006
;
107
:
190
6
.
21.
Rubenstein
JL
,
Fridlyand
J
,
Shen
A
,
Aldape
K
,
Ginzinger
D
,
Batchelor
T
, et al
Gene expression and angiotropism in primary CNS lymphoma
.
Blood
2006
;
107
:
3716
23
.
22.
Alizadeh
AA
,
Eisen
MB
,
Davis
RE
,
Ma
C
,
Lossos
IS
,
Rosenwald
A
, et al
Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling
.
Nature
2000
;
403
:
503
11
.
23.
Montesinos-Rongen
M
,
Brunn
A
,
Bentink
S
,
Basso
K
,
Lim
WK
,
Klapper
W
, et al
Gene expression profiling suggests primary central nervous system lymphomas to be derived from a late germinal center B cell
.
Leukemia
2008
;
22
:
400
5
.
24.
Tun
HW
,
Personett
D
,
Baskerville
KA
,
Menke
DM
,
Jaeckle
KA
,
Kreinest
P
, et al
Pathway analysis of primary central nervous system lymphoma
.
Blood
2008
;
111
:
3200
10
.
25.
Sung
CO
,
Kim
SC
,
Karnan
S
,
Karube
K
,
Shin
HJ
,
Nam
DH
, et al
Genomic profiling combined with gene expression profiling in primary central nervous system lymphoma
.
Blood
2011
:
117
:
1291
300
.
26.
Shipp
MA
,
Ross
KN
,
Tamayo
P
,
Weng
AP
,
Kutok
JL
,
Aguiar
RC
, et al
Diffuse large B-cell lymphoma outcome prediction by gene-expression profiling and supervised machine learning
.
Nat Med
2002
;
8
:
68
74
.
27.
Rosenwald
A
,
Wright
G
,
Chan
WC
,
Connors
JM
,
Campo
E
,
Fisher
RI
, et al
Lymphoma/Leukemia Molecular Profiling Project. The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma
.
N Engl J Med
2002
;
346
:
1937
47
.
28.
Cordell
HJ
. 
Detecting gene-gene interactions that underlie human diseases
.
Nat Rev Genet
2009
;
10
:
392
404
.
29.
Wang
MG
,
Yi
H
,
Guerini
D
,
Klee
CB
,
McBride
OW
. 
Calcineurin A alpha (PPP3CA), calcineurin A beta (PPP3CB) and calcineurin B (PPP3R1) are located on human chromosomes 4, 10q21–>q22 and 2p16–>p15 respectively
.
Cytogenet Cell Genet
1996
;
72
:
236
41
.
30.
Kennedy
RD
,
Quinn
JE
,
Johnston
PG
,
Harkin
DP
. 
BRCA1: mechanisms of inactivation and implications for management of patients
.
Lancet
2002
;
360
:
1007
14
.
31.
Kennedy
RD
,
Quinn
JE
,
Mullan
PB
,
Johnston
PG
,
Harkin
DP
. 
The role of BRCA1 in the cellular response to chemotherapy.
J Natl Cancer Inst
2004
;
96
:
1659
68
.
32.
Clark-Knowles
KV
,
O'Brien
AM
,
Weberpals
JI
. 
BRCA1 as a therapeutic target in sporadic epithelial ovarian cancer
.
J Oncol
2010
;
2010
:
891059
.
33.
Quinn
JE
,
James
CR
,
Stewart
GE
,
Mulligan
JM
,
White
P
,
Chang
GK
, et al
BRCA1 mRNA expression levels predict for overall survival in ovarian cancer after chemotherapy
.
Clin Cancer Res
2007
;
13
:
7413
20
.
34.
Rakha
EA
,
El-Sheikh
SE
,
Kandil
MA
,
El-Sayed
ME
,
Green
AR
,
Ellis
IO
. 
Expression of BRCA1 protein in breast cancer and its prognostic significance
.
Hum Pathol
2008
;
39
:
857
65
.
35.
Margeli
M
,
Cirauqui
B
,
Castella
E
,
Tapia
G
,
Costa
C
,
Gimenez-Capitan
A
, et al
The prognostic value of BRCA1 mRNA expression levels following neoadjuvant chemotherapy in breast cancer
.
PLoS ONE
2010
;
5
:
e9499
.
36.
Forentino
M
,
Judson
G
,
Penney
K
,
Flavin
R
,
Stark
J
,
Fiore
C
, et al
Immunohistochemical expression of BRCA1 and lethal prostate cancer
.
Cancer Res
2010
;
70
:
3136
9
.
37.
Taron
M
,
Rosell
R
,
Felip
E
,
Mendez
P
,
Souglakos
J
,
Ronco MS
, et al
BRCA1 mRNA expression levels as an indicator of chemoresistance in lung cancer
.
Hum Mol Genet
2004
;
13
:
2443
9
.
38.
Rosell
R
,
Skrzypski
M
,
Jassem
E
,
Taron
M
,
Bartolucci
R
,
Sanchez
JJ
, et al
BRCA1: a novel prognostic factor in resected non–small-cell lung cancer
.
PLoS ONE
2007
;
2
:
e1129
.
39.
Boukovinas
I
,
Papadaki
C
,
Mendez
P
,
Taron
M
,
Mavroudis
D
,
Koutsopoulos
A
, et al
Tumor BRCA1, RRM1 and RRM2 mRNA expression levels and clinical response to first-line gemcitabine plus docetaxel in non-small-cell lung cancer patients
.
PLoS ONE
2008
;
3
:
e3695
.
40.
Rosell
R
,
Perez-Roca
L
,
Sanchez
JJ
,
Cobo
M
,
Moran
T
,
Chaib
I
, et al
Customized treatment in non–small-cell lung cancer based on EGFR mutations and BRCA1 mRNA expression
.
PLoS ONE
2009
;
4
:
e5133
.
41.
Esteller
M
,
Silva
JM
,
Dominguez
G
,
Bonilla
F
,
Matias-Guiu
X
,
Lerma
E
, et al
Promoter hypermethylation and BRCA1 inactivation in sporadic breast and ovarian tumors
.
J Natl Cancer Inst
2000
;
92
:
564
9
.
42.
Wang
B
,
Matsuoka
S
,
Ballif
BA
,
Zhang
D
,
Smogorzewska
A
,
Gygi
SP
, et al
Abraxas and RAP80 form a BRCA1 protein complex required for the DNA damage response
.
Science
2007
;
316
:
1194
8
.
43.
Wu
M
,
Soler
DR
,
Abba
MC
,
Nunez
MI
,
Baer
R
,
Hatzis
C
, et al
CtIP silencing as a novel mechanism of tamoxifen resistance in breast cancer
.
Mol Cancer Res
2007
;
5
:
1285
95
.
44.
Soussain
C
,
Hoang-Xuan
K
,
Taillandier
L
,
Fourme
E
,
Choquet
S
,
Witz
F
, et al
Intensive chemotherapy followed by hematopoietic stem-cell rescue for refractory and recurrent primary CNS and intraocular lymphoma: Société Française de Greffe de Moëlle Osseuse-Thérapie Cellulaire
.
J Clin Oncol
2009
;
26
:
2512
8
.