16
Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018 SCIENCE TRANSLATIONAL MEDICINE | RESEARCH ARTICLE 1 of 15 BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu 1 , Giovanna M. Bernal 1 , Kirk E. Cahill 1 , Peter Pytel 2 , Carrie A. Fitzpatrick 2 , Heather Mashek 2 , Ralph R. Weichselbaum 3 , Bakhtiar Yamini 1 * The response of patients with gliomas to alkylating chemotherapy is heterogeneous. However, there are currently no universally accepted predictors of patient response to these agents. We identify the nuclear factor B (NF-B) co-regulator B cell CLL/lymphoma 3 (BCL-3) as an independent predictor of response to temozolomide (TMZ) treatment. In glioma patients with tumors that have a methylated O 6 -methylguanine DNA methyltransferase (MGMT) promoter, high BCL-3 expression was associated with a poor response to TMZ. Mechanistically, BCL-3 promoted a more malignant phenotype by inducing an epithelial-to-mesenchymal transition in glioblastomas through promoter-specific NF-B dimer exchange. Carbonic anhydrase II (CAII) was identified as a downstream factor promoting BCL-3–mediated resistance to chemotherapy. Experiments in glioma xenograft mouse models demonstrated that the CAII inhibitor acetazolamide enhanced survival of TMZ-treated animals. Our data suggest that BCL-3 might be a useful indicator of glioma response to alkylating chemotherapy and that acetazolamide might be repurposed as a chemosensitizer for treating TMZ-resistant gliomas. INTRODUCTION Diffuse gliomas encompass both lower-grade gliomas (LGGs), in- cluding grade II and III tumors, and glioblastoma (GBM). Within each grade, subgroups of tumors that have significantly different responses to therapy exist (1, 2). Despite this heterogeneity, virtually all glioma patients receive alkylating chemotherapy as part of their treatment regimen. Although alkylators like temozolomide (TMZ) improve overall patient survival (3), many patients experience mini- mal benefit from their use. These observations underline the critical need for predictors of response to alkylating therapy. Glioma cells are distinguished by the presence of recurrent copy number alterations (CNAs) affecting broad chromosomal regions (4). Although these alterations primarily involve driver genes, they can also affect nearby passenger genes not directly involved in ma- lignant progression (5). It has been suggested that targeting such flanking genes might have therapeutic effects (6). However, passenger genes, unlike drivers, have not yet been shown to mediate therapeutic susceptibility in clinical glioma (7). The best described mechanism of resistance to TMZ involves direct cytotoxic lesion repair by the DNA repair enzyme O 6 -methylguanine DNA methyltransferase (MGMT) (8). However, even in tumors with low MGMT expression, other pathways have been shown to inde- pendently modulate response to therapy and patient outcome (9). Nuclear factor B (NF-B) plays an important role in promoting resistance to DNA-damaging agents in GBM (10, 11). In addition to the five primary NF-B subunits, p50 (NF-B1, p105), p52 (NF-B2, p100), p65 (RelA), RelB, and c-Rel (12), multiple co-regulators also con- tribute to the overall downstream response. B cell CLL/lymphoma 3 (BCL-3) is an atypical inhibitor B (IB) protein that both activates and inhibits NF-B signaling (13). BCL-3 is a candidate oncoprotein shown to be up-regulated in several malignancies (14, 15) that regu- lates NF-B signaling in conjunction with p50- and p52-containing dimers (1618). Here, we show that BCL-3 reduced TMZ-induced cytotoxicity in gliomas through the activation of carbonic anhydrase II (CAII). In addition, we found that the CA inhibitor acetazolamide (ACZ) po- tentiated the effect of TMZ in mouse models of glioma, identifying a potential repurposing strategy for TMZ chemosensitization. RESULTS BCL3 is an independent predictor of response to TMZ in GBM To examine the role of BCL-3 in response to TMZ, we altered its expression in patient-derived glioma stem-like cells (GSCs) (table S1) using vectors expressing human BCL-3 or short hairpins (sh) target- ing BCL3 (fig. S1, A and B). Knockdown of BCL3 in GSCs with high basal BCL-3 increased cell death after treatment with TMZ (Fig. 1A and fig. S1C). Conversely, overexpression of BCL-3 in GSCs with low basal BCL-3 decreased TMZ-induced death (Fig. 1B and fig. S1D). Moreover, in U87, U251, and T98 GBM cell lines, knockdown of BCL3 with short interfering RNA (si-RNA) increased the TMZ- induced effect on clonal survival (fig. S1E), whereas overexpression of BCL-3 reduced the efficacy of TMZ (Fig. 1C and fig. S1F). These findings indicate that BCL-3 modulates cytotoxicity induced by TMZ. Next, to investigate BCL-3 in clinical GBM, we examined four independent expression databases. In these data sets, patients with high BCL3 expression had reduced survival compared to patients with low BCL3 expression (Fig. 1D, fig. S1G, and table S2). BCL3 expression was significantly associated with survival on multivariate analysis, taking the primary GBM prognostic factors into consider- ation [hazard ratio (HR), 1.455; P = 0.017; Table 1]. The expression of none of the primary NF-B subunits correlated with survival (fig. S1H). Similarly, NFKBIA, the gene encoding IB, a potential tumor sup- pressor in GBM (19), also had no prognostic value on its own (fig. S1I). These results indicate that BCL3 expression plays an independent role in GBM patient outcome. To understand whether BCL3 is specifically relevant in the setting of alkylation damage, we looked at TCGA to analyze whether BCL3 ex- pression affected survival of GBM patients who were untreated, treated with ionizing radiation (IR) alone, or treated with IR in combination 1 Section of Neurosurgery, Department of Surgery, University of Chicago, Chicago, IL 60637, USA. 2 Department of Pathology, University of Chicago, Chicago, IL 60637, USA. 3 Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA. *Corresponding author. Email: [email protected] Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works by guest on June 22, 2020 http://stm.sciencemag.org/ Downloaded from

BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

  • Upload
    others

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Page 1: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

1 of 15

B R A I N T U M O R S

BCL3 expression promotes resistance to alkylating chemotherapy in gliomasLongtao Wu1, Giovanna M. Bernal1, Kirk E. Cahill1, Peter Pytel2, Carrie A. Fitzpatrick2, Heather Mashek2, Ralph R. Weichselbaum3, Bakhtiar Yamini1*

The response of patients with gliomas to alkylating chemotherapy is heterogeneous. However, there are currently no universally accepted predictors of patient response to these agents. We identify the nuclear factor B (NF-B) co-regulator B cell CLL/lymphoma 3 (BCL-3) as an independent predictor of response to temozolomide (TMZ) treatment. In glioma patients with tumors that have a methylated O6-methylguanine DNA methyltransferase (MGMT) promoter, high BCL-3 expression was associated with a poor response to TMZ. Mechanistically, BCL-3 promoted a more malignant phenotype by inducing an epithelial-to-mesenchymal transition in glioblastomas through promoter-specific NF-B dimer exchange. Carbonic anhydrase II (CAII) was identified as a downstream factor promoting BCL-3–mediated resistance to chemotherapy. Experiments in glioma xenograft mouse models demonstrated that the CAII inhibitor acetazolamide enhanced survival of TMZ-treated animals. Our data suggest that BCL-3 might be a useful indicator of glioma response to alkylating chemotherapy and that acetazolamide might be repurposed as a chemosensitizer for treating TMZ-resistant gliomas.

INTRODUCTIONDiffuse gliomas encompass both lower-grade gliomas (LGGs), in-cluding grade II and III tumors, and glioblastoma (GBM). Within each grade, subgroups of tumors that have significantly different responses to therapy exist (1, 2). Despite this heterogeneity, virtually all glioma patients receive alkylating chemotherapy as part of their treatment regimen. Although alkylators like temozolomide (TMZ) improve overall patient survival (3), many patients experience mini-mal benefit from their use. These observations underline the critical need for predictors of response to alkylating therapy.

Glioma cells are distinguished by the presence of recurrent copy number alterations (CNAs) affecting broad chromosomal regions (4). Although these alterations primarily involve driver genes, they can also affect nearby passenger genes not directly involved in ma-lignant progression (5). It has been suggested that targeting such flanking genes might have therapeutic effects (6). However, passenger genes, unlike drivers, have not yet been shown to mediate therapeutic susceptibility in clinical glioma (7).

The best described mechanism of resistance to TMZ involves direct cytotoxic lesion repair by the DNA repair enzyme O6-methylguanine DNA methyltransferase (MGMT) (8). However, even in tumors with low MGMT expression, other pathways have been shown to inde-pendently modulate response to therapy and patient outcome (9).

Nuclear factor B (NF-B) plays an important role in promoting resistance to DNA-damaging agents in GBM (10, 11). In addition to the five primary NF-B subunits, p50 (NF-B1, p105), p52 (NF-B2, p100), p65 (RelA), RelB, and c-Rel (12), multiple co-regulators also con-tribute to the overall downstream response. B cell CLL/lymphoma 3 (BCL-3) is an atypical inhibitor B (IB) protein that both activates and inhibits NF-B signaling (13). BCL-3 is a candidate oncoprotein shown to be up-regulated in several malignancies (14, 15) that regu-lates NF-B signaling in conjunction with p50- and p52-containing dimers (16–18).

Here, we show that BCL-3 reduced TMZ-induced cytotoxicity in gliomas through the activation of carbonic anhydrase II (CAII). In addition, we found that the CA inhibitor acetazolamide (ACZ) po-tentiated the effect of TMZ in mouse models of glioma, identifying a potential repurposing strategy for TMZ chemosensitization.

RESULTSBCL3 is an independent predictor of response to TMZ in GBMTo examine the role of BCL-3 in response to TMZ, we altered its expression in patient-derived glioma stem-like cells (GSCs) (table S1) using vectors expressing human BCL-3 or short hairpins (sh) target-ing BCL3 (fig. S1, A and B). Knockdown of BCL3 in GSCs with high basal BCL-3 increased cell death after treatment with TMZ (Fig. 1A and fig. S1C). Conversely, overexpression of BCL-3 in GSCs with low basal BCL-3 decreased TMZ-induced death (Fig. 1B and fig. S1D). Moreover, in U87, U251, and T98 GBM cell lines, knockdown of BCL3 with short interfering RNA (si-RNA) increased the TMZ- induced effect on clonal survival (fig. S1E), whereas overexpression of BCL-3 reduced the efficacy of TMZ (Fig. 1C and fig. S1F). These findings indicate that BCL-3 modulates cytotoxicity induced by TMZ.

Next, to investigate BCL-3 in clinical GBM, we examined four independent expression databases. In these data sets, patients with high BCL3 expression had reduced survival compared to patients with low BCL3 expression (Fig. 1D, fig. S1G, and table S2). BCL3 expression was significantly associated with survival on multivariate analysis, taking the primary GBM prognostic factors into consider-ation [hazard ratio (HR), 1.455; P = 0.017; Table 1]. The expression of none of the primary NF-B subunits correlated with survival (fig. S1H). Similarly, NFKBIA, the gene encoding IB, a potential tumor sup-pressor in GBM (19), also had no prognostic value on its own (fig. S1I). These results indicate that BCL3 expression plays an independent role in GBM patient outcome.

To understand whether BCL3 is specifically relevant in the setting of alkylation damage, we looked at TCGA to analyze whether BCL3 ex-pression affected survival of GBM patients who were untreated, treated with ionizing radiation (IR) alone, or treated with IR in combination

1Section of Neurosurgery, Department of Surgery, University of Chicago, Chicago, IL 60637, USA. 2Department of Pathology, University of Chicago, Chicago, IL 60637, USA. 3Department of Radiation and Cellular Oncology and the Ludwig Center for Metastasis Research, University of Chicago, Chicago, IL 60637, USA.*Corresponding author. Email: [email protected]

Copyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement of Science. No claim to original U.S. Government Works

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 2: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

2 of 15

with TMZ. BCL3 expression did not segregate GBM patients into survival groups if they were untreated or treated with IR alone (Fig. 1E and fig. S1J). Consistent with this latter finding, overexpression of BCL-3 in GBM cells did not alter clonal survival in response to IR (fig. S1K). However, when TMZ was added to IR, patients with low BCL3 expression survived significantly longer than those with high expression (P = 0.0084; Fig. 1E and fig. S1J). In patients treated with TMZ, BCL3 expression remained associated with survival on multi-variate analysis, taking into account MGMT promoter methylation, the primary factor associated with resistance to alkylating chemotherapy in GBM (table S3) (8). Moreover, our analysis revealed that addition of TMZ improved survival only in tumors with low BCL3 expression (Fig. 1E, inset graphs). To validate this role of BCL3, we examined a second GBM expression data set, the Repository of Molecular Brain Neoplasia Data (REMBRANDT) (20). In this data set, patients also

received procarbazine, lomustine, and vincristine (PCV) as alkylating chemotherapy. In REMBRANDT, BCL3 expression was only informa-tive of survival in patients whose treatment regimen included alkylating chemotherapy (fig. S1L). These data indicate that BCL3 expression was not an intrinsic prognostic factor in GBM or informa-tive of response to IR but specifically modulated the response to alkylating chemotherapy.

Given the link between alkylation damage and MGMT promoter methylation (8), we divided patients based on MGMT promoter methylation, as assessed by the MGMT-STP27 model (21). BCL3 expression only identified distinct survival groups in tumors with high MGMT promoter methylation (Fig. 1F). Specifically, in GBM with high MGMT promoter methylation, the HR based on BCL3 expression was greater than in the entire GBM population (1.88 ver-sus 1.41, respectively, univariate Cox regression; Fig. 1F). The data

D

A

E

F

REMBRANDT GBM

P = 0.0144

1500 50 100Survival time (months)

Perc

ent s

urvi

val

50

100

0

25

75

BCL3 low(n = 90)BCL3 high(n = 90)

GSE16011

P = 0.0230

1500 50 100Survival time (months)

Perc

ent s

urvi

val

50

100

0

25

75

BCL3 low(n = 79)BCL3 high(n = 79)

G H

UChicago cohort BCL-3 IHC

1000 20 80Survival time (months)

40 60

P < 0.0001Per

cent

sur

viva

l

50

100

0

25

75BCL-3 low (n = 58)BCL-3 high (n = 28)

B C

0 60Survival time (months)

20 40

Per

cent

sur

viva

l

50

100

0

25

75IR & TMZ/BCL3 low

IR only/BCL3 low

IR & TMZ/BCL3 high

No treatment/BCL3 highNo treatment/BCL3 low

IR only/BCL3 high

TCGA GBM

P = 0.516IR & TMZ (n = 143)

IR only (n = 43)

0 20 60Survival time (months)

40

Per

cent

sur

viva

l

50

100

0

25

75

IR & TMZ (n = 142)

IR only (n = 42)

P = 0.034

0 20 60Survival time (months)

40

Per

cent

sur

viva

l50

100

0

25

75

BCL-3 low

Pat

ient

1

BCL-3 high

Pat

ient

2

Pat

ient

3P

atie

nt 4

BCL3 low

BCL3 High

HR = 1.595(CI, 1.092 2.329)

HR = 1.479(CI, 1.084 2.069)HR = 1.606

(CI, 1.201 2.148)

HR = 0.8826(CI, 0.605 1.287)

HR = 0.6240(CI, 0.403 0.964)

HR = 5.418(CI, 2.747 9.647)

P = 0.0014

1500 50 100Survival time (months)

TCGA GBM

Perc

ent s

urvi

val

50

100

0

25

75

BCL3 low(n = 127)BCL3 high(n = 127)

0 50 100 150

0

25

50

75

100

Perc

ent s

urvi

val

HR = 1.31CI = 0.91 1.88

HR = 1.87CI =1.20 2.91

BCL3 lowMGMT unmethylated

BCL3 low

BCL3 high

BCL3 high

P = 0.40

P = 0.013

(n = 86)(n = 87)

(n = 83)(n = 83)

MGMT methylated

Survival time (months)

0.0

1.0

2.0

3.0

4.0

5.0

Rel

ativ

e ce

ll de

ath BCL-3

GAPDH

EV BCL-3

**

****

- 50 100 200TMZ (µM)0.0

0.2

0.4

0.6

0.8

1.0

1.2

Sur

vivi

ng fr

actio

n

EV BCL-3

BCL-3GAPDH

EV BCL-3

*

*

*

- 25 50 75TMZ (µM)

U87 (BCL-3 intermediate)

0.01.02.03.04.05.06.07.0

Rel

ativ

e ce

ll de

ath

BCL-3GAPDH

- 25 50 100TMZ (µM)

****

sh-SCR sh-BCL-3 #1 sh-BCL-3 #2

GBM34 (BCL-3 high)

sh-B

CL3 #2

sh-S

CR

sh-B

CL3 #1

EV BCL-3

GBM44 (BCL-3 low)

Rel

ativ

e ce

ll de

ath

BCL-3GAPDH

EV BCL-3**

- 200TMZ (µM)

EV BCL-3

GBM8 (BCL-3 low)

****

Fig. 1. BCL3 predicts response to alkylating chemotherapy in GBM. (A and B) Trypan blue assays in GSCs expressing the indicated construct treated with TMZ (72 hours). Data show fold change in percent dead cells relative to cells expressing a scrambled RNA sequence (sh-SCR), or empty vector (EV), treated with vehicle, ± SD (n = 3). Inset: Immunoblotting (IB) with anti–BCL-3. (C) Clonogenic assay in U87 cells stably expressing BCL-3 or EV treated with vehicle or TMZ. Data show mean value relative to vehicle ± SD (n = 3). GAPDH, glyceraldehyde-3-phosphate dehydrogenase. (D to F) Kaplan-Meier overall survival curves in GBM patients. (D) Survival based on median mRNA expression (REMBRANDT and GSE16011) and upper and lower quartile (TCGA). (E) TCGA GBM patients separated by median BCL3 expression and treatment modality. n = 490 patients total: no treatment, IR alone, and IR + TMZ; inset graphs: IR versus IR + TMZ in patients with high or low BCL3 mRNA (median cutoff). (F) TCGA GBM patients separated by median MGMT promoter methylation and median BCL3 expression. (G) Representative IHC staining for BCL-3 in GBM. (H) Survival curves in GBM patients separated by BCL-3 IHC staining. Kaplan-Meier curves analyzed by the log-rank test. *P < 0.05 and **P < 0.01.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 3: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

3 of 15

showed that the prognosis of patients with MGMT promoter meth-ylated tumors that also had high BCL3 expression was similar to that in patients with an unmethylated MGMT promoter (Fig. 1F).

As public data sets primarily examine mRNA, to study the role of BCL-3 protein, we obtained specimens from a consecutive series of glioma patients. Individual slides and tissue microarrays (TMAs) were established, and BCL-3 protein was examined by immunohis-tochemistry (IHC) in 86 confirmed GBMs. IHC grading was per-formed in a blinded fashion based on a four-tier system that was converted into a binary scale (Fig. 1G). Clinical characteristics are noted (table S4); greater than 90% of patients received TMZ. Data analysis showed that GBM patients with low BCL-3 staining sur-vived significantly longer than those with high staining [P < 0.0001; HR, 5.418; 95% confidence interval (CI), 2.747 to 9.647; Fig. 1H], a finding independent of age. These results support the mRNA data and emphasize the importance of BCL-3 protein in modulating the response to alkylation damage.

BCL3 is predictive in all diffuse gliomasAlkylating chemotherapy is also important in the management of LGGs. Examination of multiple LGG data sets demonstrated that BCL3 expression separated these patients into distinct survival groups (Fig. 2A, fig. S2A, and table S5). In addition, our analysis showed that there was no difference in BCL3 expression between grade II and III tumors (fig. S2, B and C). Multivariate analysis that included age, isocitrate dehydrogenase 1 (IDH1) mutation, 1p/19q co-deletion, and MGMT promoter methylation showed that BCL3 expression remained associated with survival (Table 2); moreover, BCL3 separated LGG patients into different survival groups in tumors with high MGMT promoter methylation (Fig. 2B). We next parti-tioned patients by treatment modality. Consistent with a predictive effect, BCL3 expression was not informative in LGG patients who did not receive an alkylating agent or those treated with IR alone, whereas in patients treated with TMZ (either alone or with PCV) in combination with IR, BCL3 expression identified distinct survival groups (Fig. 2C). When data from patients treated with alkylating chemotherapy were analyzed on multivariate analysis, low numbers of patients achieving the endpoint of death precluded meaningful statis-tical analysis (table S6). To validate the predictive nature of BCL3, we examined LGGs from REMBRANDT. In this data set, BCL3 ex-pression was informative of patient survival only in LGG patients who received alkylating chemotherapy, of which >95% included TMZ (fig. S2D). In addition, given the importance of randomized studies for identifying

predictive factors, we looked at European Organization for Research and Treatment of Cancer (EORTC) study 26951, a randomized phase III clinical trial that examined addition of alkylating chemotherapy to IR in LGG (22). In this study, performed before TMZ was routinely used, BCL3 expression was not informative of survival time in patients treated with IR alone. However, among patients who received alkylat-ing chemotherapy in addition to IR, those with low BCL3 expression survived significantly longer (HR, 2.051; P < 0.044; Fig. 2D). Moreover, multivariate analysis among patients that received chemotherapy showed that BCL3 expression remained significant (P = 0.006) when factoring in age and 1p/19q status (table S7; IDH1 mutation was not included due to incomplete patient data). These findings indicate that BCL3 is an in-dependent predictor of response to alkylating chemotherapy in LGG.

Given that IDH mutation status is as important a prognostic marker as tumor grade (23), we examined a pan-glioma data set (grade II, III, and IV tumors) in which patients were separated by IDH1 mu-tation status (24). Notably, IDH1-mutant (IDH-mt) gliomas had lower BCL3 expression than IDH–wild type (wt) (Fig. 2E). Similar results were seen with CpG island methylator phenotype (CIMP)–positive tumors compared to CIMP-negative tumors (fig. S2E). Our analysis also showed that BCL3 expression was reduced in LGG com-pared to GBM (P < 0.001, fig. S2F). In this pan-glioma data set, BCL3 was correlated with survival on multivariate analysis, taking IDH1 mutation, MGMT methylation, 1p/19q co-deletion, and age into con-sideration (Table 3). Consistent with a predictive effect, BCL3 ex-pression separated pan-glioma patients into distinct survival groups only if they received alkylating chemotherapy in their treatment regi-men (Fig. 2F). In these patients, BCL3 expression remained significant (P = 0.023) on multivariate analysis incorporating IDH1 mutation and MGMT promoter methylation (table S8). In summary, these data indicate that BCL3 is an independent predictor of response to al-kylating chemotherapy in diffuse gliomas.

BCL3 loss is a passenger event associated with survivalMultiple transcription factors can contribute to BCL3 mRNA ex-pression (13); however, it is notable that BCL3 is found on 19q13, a chromosomal band deleted in 20 to 40% of all types of glioma (25–29). We therefore examined the relationship between BCL3 copy number (CN) and expression. In both LGGs and GBMs, tumors with BCL3 CN loss had significantly lower BCL3 mRNA expression (P < 0.003; Fig. 3A). Moreover, in an independent data set, LGGs and GBM with 19q deletion had significantly lower BCL3 expression than nonde-leted tumors (P < 0.01; Fig. 3B). To examine the link between CN

Table 1. Cox regression analysis in TCGA GBM patients.

CovariateUnivariate regression Multivariate regression

HR 95% CI P HR 95% CI P

BCL3 expression 1.408 1.131–1.752 0.002 1.455 1.069–1.981 0.017

MGMT methylation 0.624 0.482–0.808 0.000 0.754 0.580–0.981 0.035

IDH1 mutation 0.232 0.075–0.724 0.012 0.490 0.153–1.573 0.231

Radiotherapy 0.357 0.287–0.445 0.000 0.424 0.307–0.585 0.000

Chemotherapy 0.576 0.471–0.705 0.000 0.650 0.492–0.860 0.003

Procurement method 1.155 0.864–1.543 0.330 1.055 0.734–1.516 0.773

Age 1.035 1.027–1.044 0.000 1.027 1.016–1.039 0.000

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 4: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

4 of 15

and protein expression, we performed fluorescence in situ hybrid-ization (FISH) on TMA samples. Tumors with hemizygous BCL3 deletion had significantly lower BCL-3 than nondeleted tumors, as assessed by IHC (P = 0.0007; Fig. 3C). The association between CN and BCL3 expression raised the question of whether CN is also linked to survival. In both LGG and GBM groups, patients who had tumors with BCL3 deletion survived significantly longer than those with non-deleted tumors (P < 0.05; Fig. 3D). Moreover, BCL3 CN remained significantly associated with survival on multivariate analysis (P = 0.021; table S9). These results indicate that genetic deletion of BCL3 is as-sociated with reduced BCL-3 expression and improved survival.

The prevalence of BCL3 loss suggests either that BCL3 is a driver of glioma formation or that it is inadvertently affected by alterations targeted to a large chromosomal region. The lack of reported BCL3 mutations or translocations in glioma suggested that BCL3 loss is not a driver of these tumors (fig. S3A). Conversely, CNAs of BCL3 cor-related with those of nearby genes (fig. S3B), suggesting that their CNAs are closely linked. Therefore, we examined the CN status of

the genes surrounding BCL3 in 19q13.31-32. Analysis of TCGA data showed that in both GBM and LGG, loss of all the genes in this band correlated not only with BCL3 but also with the other genes in the region (Fig. 3E and fig. S3C), suggesting that BCL3 deletion occurs as a result of modifications targeted to a broad region of 19q13. This finding is consistent not only with the fact that the entire 19q arm is lost in oligodendrogliomas but also with the observation that, in as-trocytic tumors and GBM, CNAs within 19q13 involve a broad re-gion (26, 30). The above data support the hypothesis that BCL3 loss is a passenger event seen in all types of glioma.

Despite the importance of deletion, some BCL3 nondeleted gliomas also had low mRNA expression. Given the role of epigenetic modi-fications in regulating expression, we examined BCL3 promoter meth-ylation. In TCGA pan-glioma data, a significant inverse correlation between BCL3 promoter methylation and expression was seen (P < 0.0001; fig. S3D). Although the correlation was evident in IDH-wt and IDH-mt tumors, this was stronger in IDH-wt tumors (fig. S3D). Analysis of the pan-glioma data set demonstrated that BCL3 promoter methylation

TCGA LGG

P < 0.0001

1500 50 100Survival time (months)

250200

BCL3 low(n = 54)BCL3 high(n = 55)

Per

cent

sur

viva

l

50

100

0

25

75

B

C

A

FE

P = 0.3516

BCL3 low(n = 20)BCL3 high(n = 21)

Per

cent

sur

viva

l50

100

0

25

75

1500 50 100Survival time (months)

No chemotherapyTCGA LGG

GSE16011 (LGG)

TCGA LGGIR only

P = 0.6865

BCL3 low(n = 19)

BCL3 high(n = 19)P

erce

nt s

urvi

val

50

100

00 40 100Survival time (months)

806020

1500 50 100Survival time (months)

Per

cent

sur

viva

l

50

100

0

25

75

P = 0.0053

BCL3 low(n = 247)BCL3 high(n = 248)

TMZ at any time

P = 0.0030

BCL3 low(n = 130)BCL3 high(n = 129)

Per

cent

sur

viva

l

50

100

0

25

75

1500 50 100Survival time (months)

TCGA LGG

HR = 2.386(CI, 1.530 3.719)

HR = 2.202(CI, 1.322 3.667)

Survival time (months)

Per

cent

sur

viva

l

0 50 100 1500

25

50

75

100

P = 0.0833

P = 0.0332

BCL3 lowMGMT unmethylated

BCL3 low

BCL3 high

BCL3 high

MGMT Methylated

(n = 42)(n = 43)

(n = 204)(n = 203)

0 50 100Survival time (months)

0

25

50

75

100

Per

cent

sur

viva

l

BCL3 highBCL3 low

BCL3 lowBCL3 high P = 0.665

P < 0.0001

Pan-glioma

+ Alkalyator

No Alkalyator (n = 66)(n = 67)

(n = 161)(n = 161)

12

6

8

4

P < 0.0001

TCGA

wt mtIDH

10

Nor

mal

ized

BC

L3 e

xpre

ssio

n

GSE1601112

6

8

4

P < 0.0001

wt mtIDH

10

Nor

mal

ized

BC

L3 e

xpre

ssio

n

Pan-glioma

TCGA LGG

D

IR only

Perc

ent s

urviv

al

50

100

0

25

75

Perc

ent s

urviv

al

50

100

0

25

75

Survival time (months)0 20 40 60 80 100

Survival time (months)0 20 40 60 80 100

P = 0.044

BCL3 low(n = 24)BCL3 high(n = 25)

P = 0.519

BCL3 low(n = 23)BCL3 high(n = 23)

IR + alkylator

HR = 2.051CI, 1.018 4.129

HR = 1.228CI, 0.658 2.289

EORTC 26951(GSE43107)

Fig. 2. BCL3 is relevant in LGGs and pan-glioma patients. (A and B) Survival curves from (A) LGG data sets separated by median BCL3 expression and (B) TCGA LGG patients separated by median BCL3 expression and median MGMT promoter methylation. (C) Survival curves in TCGA LGG patients based on median BCL3 expression and treatment (n = 338 patients total). For patients treated with TMZ, 202 received only TMZ, and 57 received TMZ and PCV. No chemotherapy group includes patients with and without IR. IR alone patients received no alkylating agents. (D) Survival curves in EORTC 26951 based on median BCL3 expression. Patients were separated by treat-ment modality into those treated with IR alone versus IR + alkylating chemotherapy. (E) BCL3 mRNA expression in pan-glioma patients based on IDH1 mutation status in the indicated data set. For TCGA: IDH1-wt, n = 184; IDH1-mt, n = 406. For GSE16011: IDH1-wt, n = 143; IDH1-mt, n = 83. (F) Survival curves in TCGA pan-glioma patients separated by median BCL3 expression and by alkylating chemotherapy treatment (yes/no). Kaplan-Meier curves analyzed by the log-rank test.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 5: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

5 of 15

was significantly associated with survival on multivariate analysis in-corporating IDH mutation, MGMT promoter methylation, and 1p/19q co-deletion (P = 0.018; table S10). Moreover, when the data were sepa-rated by the patients’ IDH status, BCL3 promoter methylation was only informative in patients with IDH-wt tumors (Fig. 3F), a finding consistent with the stronger correlation between BCL3 methylation and expression in IDH-wt compared to IDH-mt tumors (fig. S3D). These results indicate that, in glioma, the genetic and epigenetic al-terations of BCL3 that regulate expression are significantly associated with survival.

Given the correlation between BCL3 CN loss and that of other 19q13 genes, we examined whether the expression of other 19q13 genes was also associated with survival. Unlike BCL3, the mRNA expres-sion of none of the surrounding 19q13 genes (fig. S3C) could sepa-rate TCGA GBM patients into significantly distinct survival groups, including the expression of RELB, an NF-B subunit located on 19q13 whose CNAs are tightly linked to BCL3 and have been previously associated with GBM (fig. S1H) (2, 31) (GlioVis data portal for visual-ization and analysis of brain tumor expression data sets) (32). These findings indicate that the correlation between BCL3 loss and survival does not extrapolate to all genes in the 19q13 chromosomal region.

BCL-3 induces epithelial-to-mesenchymal transition in GBMTo understand how BCL-3 promotes resistance to alkylation damage, we modulated its expression in GSCs and GBM cell lines. In A172 cells that have extremely low basal BCL-3, overexpression of BCL-3 for 2 weeks changed the cell morphology to a spindle-like appearance,

suggestive of epithelial-to-mesenchymal transition (EMT) (Fig. 4A). To further study this finding, we examined GSCs with low and high basal BCL-3 expression. Overexpression of BCL-3 in GBM44 GSCs and A172 cells increased the expression of several mesenchymal markers, including vimentin (VIM), SNAI1, and TWIST1, and re-duced the expression of the epithelial marker E-cadherin (CDH1) (Fig. 4B and fig. S4A). Conversely, knockdown of BCL3 in GSCs ex-pressing high basal BCL-3, GBM43S and GBM34, resulted in de-creased mesenchymal marker expression (Fig. 4B). One prominent feature of mesenchymal differentiation is acquisition of a more stem cell–like phenotype (1, 33) that can be demonstrated in vitro by anal-ysis of neurosphere formation. Loss of BCL3 reduced neurosphere formation ability in GBM34 cells, whereas overexpression of BCL-3 had the opposite effect in GBM44 GSCs, supporting the role of BCL-3 in mesenchymal differentiation (Fig. 4C). Consistent with the above, in TCGA GBM and LGG, a correlation between the expression of BCL3 and multiple EMT markers was seen (Fig. 4D and fig. S4B). Moreover, mesenchymal (MES) GBMs, as determined by the Phillips or Verhaak classification systems (1, 2), had significantly higher BCL3 expression than proneural (PN) tumors (P < 0.005; Fig. 4E and fig. S4C). Correlation of BCL3 with the Phillips MES or PN signature genes (1) demonstrated that, although there was a strong positive correlation between BCL3 and all the MES signature genes (mean correlation is 0.59, ±0.13; table S11), the correlation between BCL3 and PN signa-ture genes was negative (mean correlation is −0.39, ±0.17; table S11) (GlioVis data portal for visualization and analysis of brain tumor expression data sets) (32). These data suggest that, in glioma, BCL-3 promotes mesenchymal differentiation.

BCL-3 induces mesenchymal differentiation via promoter-specific NF-B dimer exchangeBCL-3 has no known enzymatic function and mediates its effects by regulating downstream gene expression. To understand how BCL-3 promotes mesenchymal differentiation, we examined genome-wide mRNA expression in GBM cells expressing si-BCL3 compared to si-control, a nontargeted sequence (table S12). As BCL-3 primarily acts by modulating NF-B signaling, we looked at NF-B–dependent factors previously identified as being specific to the MES phenotype (10). Knockdown of BCL3 caused a decrease of more than 80% of NF-B–dependent MES genes (Fig. 5A). Among the transcripts most significantly down-regulated (>2-fold, adjusted P < 0.001) were CD44, chemokine (C-C motif) ligand 2 (CCL2), colony-stimulating factor 2 (CSF2), NF-B–dependent factors critical for mesenchymal differ-entiation (10), and leukemia inhibitory factor (LIF), a primary MES signature gene (1). The BCL-3 dependence of these and several other MES genes was verified after depletion and overexpression of BCL-3 in GSCs and GBM cells (Fig. 5B and fig. S5A). In addition, in clinical GBM, a strong correlation between the expression of these factors and BCL3 was seen (Fig. 5C).

To study the mechanism by which BCL-3 modulates NF-B, we first examined an NF-B–dependent luciferase reporter. Overexpres-sion of BCL-3 in GBM cells caused an increase in basal and tumor necrosis factor– (TNF)–induced NF-B activity (Fig. 5D) and an increase in TNF-induced CD44 expression (fig. S5B), a finding pre-viously reported to promote mesenchymal differentiation (10). We then looked at the primary NF-B subunits p50, p52, and p65. Al-though overexpression of BCL-3 did not induce a change in subunit expression, it increased p65 nuclear translocation (Fig. 5E). To ex-amine this finding further, we depleted BCL3 in GBM34 GSCs.

Table 2. Cox regression analysis in TCGA LGG patients. BCL3 expression is also significant with inclusion of grade into analysis (P = 0.042).

CovariateUnivariate regression

HR 95% CI P

BCL3 expression 1.431 1.186–1.735 0.000

MGMT methylation

0.268 0.167–0.431 0.000

IDH1 mutation 0.103 0.062–0.173 0.000

1p/19q co-deletion

0.486 0.281–0.837 0.009

Grade (II or III) 3.554 2.194–5.758 0.000

Radiotherapy 0.404 0.202–0.807 0.010

Chemotherapy 0.219 0.068–0.706 0.011

Procurement method

1.402 0.911–2.159 0.125

Age 1.060 1.042–1.079 0.000

CovariateMultivariate regression

HR 95% CI P

BCL3 expression 1.374 1.096–1.722 0.006

MGMT methylation

0.685 0.363–1.293 0.243

IDH1 mutation 0.401 0.186–0.864 0.020

1p/19q co-deletion

0.646 0.339–1.229 0.183

Age 1.064 1.042–1.086 0.000

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 6: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

6 of 15

Knockdown of BCL3 using two distinct sh-RNA constructs decreased nuclear and increased cytoplasmic p65 (Fig. 5F). Given this finding, to study whether inherent differences in BCL-3 correlated with dif-ferences in nuclear p65, we first examined GSCs and GBM cell lines. In the cell lines examined, we found a positive correlation between BCL-3 and nuclear p65 protein (Fig. 5G). In addition, in clinical GBM specimens, we found a significant positive correlation between nu-clear IHC staining of BCL-3 and p65 (P = 0.009; Fig. 5H). These results suggest that, in GBM, BCL-3 induces NF-B activity and promotes p65 nuclear translocation.

Although BCL-3 did not alter the amount of nuclear p50 or p52, we examined whether it promoted MES differentiation by altering the chromatin recruitment of these subunits. We performed quantitative chromatin immunoprecipitation (qChIP) using primers spanning the B sites of several BCL-3–regulated MES genes. Overexpression of BCL-3 induced recruitment of p65 to the B sites of MES-specific genes, in-cluding CD44 and LIF (Fig. 5I and fig. S5C). Also, whereas p50 recruit-ment decreased at CD44, LIF, and CCL2 promoters, the recruitment of p52 increased (Fig. 5I and fig. S5C). Gel shift studies using probes corresponding to the CD44 and CCL2 B sites showed that, in the presence of high BCL-3, p50 DNA binding was lost, whereas p52 binding was induced at both B sites (Fig. 5J and fig. S5D). Finally, we exam-ined endogenous chromatin enrichment of NF-B subunits at MES promoters in patient-derived GSCs. The predominant subunits bound to MES promoters in GBM34 GSCs (BCL-3 high) were p65 and p52 (Fig. 5K). These findings indicate that high BCL-3 promotes MES gene expression by inducing promoter-specific NF-B dimer exchange.

The increased recruitment of p52 to MES promoters suggested that p52 is involved in mediating the MES differentiation induced by BCL-3. We therefore depleted p52/NFKB2 in GBM34 GSCs (BCL-3 high). Knockdown of p52/NFKB2 resulted in decreased expression of CD44, CCL2, and interleukin-8 (IL8) (fig. S5E). Moreover, in TCGA GBMs, there was a positive correlation between the expression of NFKB2 and these MES genes (fig. S5F). In addition, depletion of p52/NFKB2 not only attenuated basal NF-B–regulated MES gene expression but also blocked the increase induced by BCL-3 overex-pression (fig. S5G). These results indicate that p52 mediates BCL-3– dependent increase in MES gene expression.

CAII mediates BCL-3–dependent resistance to TMZThe results indicate that BCL-3 is informative specifically in patients treated with alkylating agents such as TMZ. To identify BCL-3–regulated factors that could be responsible for the resistance to TMZ and that could potentially be targeted for inhibition, we examined the gene expression response induced by TMZ, focusing on genes up-regulated by treatment. To screen for differentially expressed transcripts, we

depleted BCL3 in U87 cells, a cell line with moderate BCL-3 expression that was sensitized to TMZ by BCL3 knockdown. The sole transcript induced by TMZ in the presence of BCL-3 and down-regulated with depletion of BCL3 was CAII (Fig. 6A and tables S12 and S13). CAII was induced by TMZ in GSCs (Fig. 6B and fig. S6A), and BCL-3 was required for this induction (Fig. 6C). Also, in clinical databases, tumors with low BCL3 expression had lower CAII mRNA expression than those with high BCL3 expression, as did tumors with 19q deletion compared to the nondeleted group (Fig. 6D and fig. S6, B and C). In addition, MES GBMs had higher CAII expression than PN tumors (fig. S6D). To examine the relevance of alkyl adducts for induction of CAII by TMZ, we modulated MGMT expression. Knockdown of MGMT in GBM26 GSCs (that express high MGMT and BCL-3) in-creased TMZ-induced CAII expression (Fig. 6E), whereas expression of MGMT in GBM cells with no MGMT blocked the effect of TMZ on CAII expression (fig. S6E). These findings suggest that TMZ induces CAII via formation of cytotoxic O6-methylguanine adducts.

No CA isoforms have yet been reported to be NF-B target genes. Although knockdown of either p50/NFB1 or p65 did not alter basal CAII expression, loss of p52/NFB2 attenuated basal and TMZ-induced CAII expression in GBM34 GSCs (Fig. 6F and fig. S6F). Analysis of the CAII promoter and the upstream coding region revealed three potential B sites (B1, B2, and B3; Fig. 6G). ChIP studies demon-strated that p50, p52, and p65 are recruited to B1 and B2, but not B3, and gel shift analysis showed that NF-B binds the B2 probe (fig. S6G). In U87 cells, TMZ decreased enrichment of p50 at B1 and B2, increased enrichment of p52 (Fig. 6H), and had no effect on either BCL-3 or p65 (fig. S6H). Consistent with this, gel shift studies showed that, in response to TMZ, p52 replaced p50 in the NF-B dimer bound to the B2 probe (Fig. 6I). The above findings suggested that TMZ modulates the interaction of BCL-3 with p50 and p52. To examine this more closely, we studied GSCs with high BCL-3 ex-pression. Western blot analysis revealed that TMZ reduced the in-teraction of BCL-3 with p50 and enhanced the association of BCL-3 with p52 (Fig. 6J). Given that TMZ induces phosphorylation of p50 S329 (11) and that S329 is a residue that potentially interacts with BCL-3 (34), we examined the role of this residue in regulating the BCL-3/p50 interaction in response to TMZ. Mutation of S329 to an unphosphorylatable form blocked the dissociation of p50 and BCL-3 after TMZ treatment (Fig. 6K). Together, these findings highlight the importance of the reciprocal interaction of BCL-3 with p50 and p52 for induction of CAII by TMZ.

Finally, to examine whether CAII is involved in the cytotoxicity induced by TMZ, we altered its expression in GSCs and GBM cells. Overexpression of CAII attenuated cell death induced by TMZ (Fig. 6L and fig. S6, I and J), whereas knockdown of CAII in GBM26 GSCs led

Table 3. Cox regression analysis in TCGA pan-glioma patients. Only patients with RNA-seq data included.

CovariateUnivariate regression Multivariate regression

HR 95% CI P HR 95% CI P

BCL3 expression 2.390 2.065–2.765 0.000 1.467 1.213–1.774 0.000

MGMT methylation 0.227 0.168–0.307 0.000 0.590 0.416–0.835 0.003

IDH1 mutation 0.073 0.051–0.103 0.000 0.309 0.176–0.541 0.000

1p/19q co-deletion 0.212 0.127–0.353 0.000 0.705 0.379–1.312 0.270

Age 1.070 1.058–1.080 0.000 1.052 1.037–1.066 0.000

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 7: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

7 of 15

to an increase in TMZ-induced cytotoxicity (Fig. 6M). Moreover, depletion of CAII and BCL3 together did not increase the anti-glioma effect of TMZ any more than knockdown of BCL-3 alone (Fig. 6N), supporting the contention that CAII and BCL-3 act in the same path-way in response to TMZ. These findings indicate that BCL-3 promotes resistance to TMZ by induction of CAII as a result of promoter-specific NF-B dimer exchange.

ACZ enhances the anti-glioma effect of TMZCAII is potently blocked by the CA inhibitor ACZ (35). ACZ has been reported to be effective against GBM cells (36, 37) and to enhance apoptosis by TMZ (38, 39). Consistent with this observation, we found that although ACZ alone did not alter overall clonogenic survival of GBM cells, it enhanced the ability of TMZ to reduce survival (fig. S7A).

To study the role of BCL-3 in this response, patient-derived GSCs were examined. ACZ enhanced cytotoxicity by TMZ in GBM34 GSCs that express high BCL-3, and knockdown of BCL3 blocked the chemo-sensitizing effect of ACZ (Fig. 7A). In GBM44 GSCs that have low BCL-3 expression, ACZ did not modulate the cytotoxic effect induced by TMZ; however, overexpression of BCL-3 enabled ACZ to enhance cell death by TMZ (Fig. 7B). Similarly, depletion of BCL3 attenuated the chemosensitizing effect of ACZ on clonogenic assay (fig. S7B). These results indicate that BCL-3 is required for sensitization to TMZ by ACZ.

Although ACZ is possibly the most commonly used CA inhibitor (40), many other CA inhibitors are available that can potentially en-hance the efficacy of TMZ (41). We examined two other clinically used general CA inhibitors, methazolamide (MZM) and topiramate (TPM).

B

D

A CIHC (BCL-3) FISH (BCL3 & 19q)

Non

-del

Del

Fisher’s exact test P = 0.0007

IHC low IHC high

Del 15 0Non-del 16 15

BCL-3

BCL3

Nor

mal

ized

BC

L3

exp

ress

ion

Del

Diploid

8

Gain5

7

6

9

10

11

12

TCGA GBMTCGA LGG

Nor

mal

ized

BC

L3

exp

ress

ion

8

5

7

6

9

10

11

4

Del

Diploid Gain

****

**

P = 0.0267

1500 50 100Survival time (months)

TCGA GBM

Per

cent

sur

viva

l

50

100

0

25

75

BCL3 del(n = 60)BCL3 non-del(n = 265)

1500 50 100Survival time (months)

Per

cent

sur

viva

l

50

100

0

25

75

P = 0.0105

TCGA LGG

BCL3 del(n = 235)BCL3 non-del(n = 245)

E

GBM

8

6

10

4Nor

mal

ized

BC

L3

exp

ress

ion

P = 0.0094

19qnon-del

19qdelnon-del

19q19qdel

Nor

mal

ized

BC

L3

exp

ress

ion

8

5

7

6

9

4P = 0.0027

GSE16011LGG

F

BCL3 methylation high

BCL3 methylation low

BCL3 methylation low

BCL3 methylation high P < 0.0001

P = 0.3647

0

25

50

75

100

0 50 100 150

Per

cent

sur

viva

l

Survival time (months)

IDH-wt

IDH-mt

TCGA pan-glioma (n = 94)

(n = 94)

(n = 204)

(n = 205)

Chr19

BC

L3

42,917,858 47,484,28219,900,913 24,098,980

19p12 19q13.31-32

1.0

0.8

0.6

0.4

0.2

0

0.2

0.4

0.6

0.8

1.0

(r)BCL3 -

Fig. 3. Loss of BCL3 is a passenger event associated with improved survival. (A) Relationship between BCL3 expression [on RNA sequencing (RNA-seq)] and BCL3 CN in TCGA LGG and GBM. In LGG, del: n = 169; diploid: n = 155; gain: n = 16. In GBM, del: n = 17; diploid: n = 89; gain: n = 39. (B) BCL3 expression in LGG and GBM tumors in relation to tumor 19q deletion status [determined by microsatellite polymerase chain reaction (PCR)]. LGG: del, n = 40; non-del, n = 33. GBM: del, n = 12; non-del, n = 58. (C) Representative FISH images using BCL3 and 19q specific probes (left) and BCL-3 IHC staining (high and low, right) in human gliomas. Table shows number of tumors in each category, two-sided Fisher’s exact test. (D) Kaplan-Meier curves in LGG and GBM patients based on BCL3 CN (with and without deletion). (E) Correlogram showing cor-relation (r, y axis) between CN of genes from the indicated regions of chromosome 19 (Chr19) in n = 514 TCGA LGG patients. For 19q13.31-32 (Chr19: 42,917,858 to 47,484,282), there are 171 genes in a span of 4.5 Mbp (million base pairs); 139 of these genes have CN data available for analysis. The nearest 19p band (19p12) was used as a reference. For 19p12 (Chr19: 19,900,913 to 24,098,980), there are 59 genes in the 4.2-Mbp region, of which 36 have CN data. Location of BCL3 shown. (F) Survival in TCGA pan-glioma patients based on median BCL3 promoter methylation and IDH1 mutation status. Pearson coefficient (r) analyzed by two-sided Student’s t test. *P < 0.003 and **P < 0.0002.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 8: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

8 of 15

MZM enhanced cytotoxicity by TMZ in a BCL-3–dependent manner (fig. S7C), whereas the anti-epileptic agent TPM did not (fig. S7D).

Given that ACZ also acts on other CAs, albeit much less effec-tively (35), we also examined the requirement of CAII for the com-bination effect of ACZ and TMZ. Knockdown of CAII with si-RNA blocked the chemosensitizing effect of ACZ on TMZ (Fig. 7C), and overexpression of CAII in cells with low basal BCL-3 promoted chemo-sensitization by ACZ (fig. S7E). Moreover, in U251 GBM cells that do not express CAII, ACZ only chemosensitized these cells to TMZ when CAII was exogenously expressed (Fig. 7, D and E). The efficacy of ACZ in combination with TMZ raised the question of whether ACZ modified NF-B activity and mesenchymal differentiation. ACZ did not affect TMZ-induced NF-B inhibitory activity (fig. S7F), and the addition of ACZ also did not induce p65 phosphorylation or alter MES gene expression (fig. S7, G and H).

To examine combination TMZ and ACZ in vivo, we performed intracranial xenograft studies in mice. In a pilot study, U87 tumors were established and ACZ was administered for 10 days after initi-ation of TMZ treatment. This combination regimen did not result in a significant change in survival. However, 10 days after TMZ ini-tiation, CAII expression was still elevated (fig. S7I), suggesting that ACZ needed to be administered for a longer period of time. We therefore altered our protocol to treat animals with daily ACZ for a total of 21 days. Using this regimen, ACZ significantly increased survival time in com-bination with TMZ compared to TMZ alone (P < 0.05; Fig. 7F and fig. S7J). ACZ alone did not affect survival in this model (Fig. 7F).

Next, we replicated the experiments in more clinically relevant patient-derived xenograft (PDX) models. In GBM34 and GBM43S

xenografts that have high BCL-3 expression, addition of ACZ signifi-cantly increased survival compared to TMZ alone (P < 0.05; Fig. 7, G and H). Addition of ACZ to TMZ in GBM43S xenografts resulted in long-term survival of several animals, a finding repeatedly seen in multiple independent experiments (Fig. 7G and fig. S7L). However, in GBM26 PDX that has high MGMT expression, addition of ACZ had no chemosensitizing effect on TMZ despite the presence of BCL-3 in this tumor (fig. S7, K and M). Finally, to specifically examine the requirement of BCL-3 for chemosensitization by ACZ in vivo, we depleted BCL3 in GBM34 tumors. Whereas expression of a control sh-RNA did not modulate the response in GBM34 tumors, knock-down of BCL3 completely blocked the ability of ACZ to increase the prosurvival effect of TMZ (Fig. 7I). Together, these data indicate that ACZ, a commonly used CA inhibitor that has no antitumor effect in GBM by itself, sensitized GBM to TMZ in a BCL-3–dependent manner.

DISCUSSIONThis work identifies BCL-3 as an indicator of sensitivity to alkylating chemotherapy in GBM. Although BCL3 expression separated all GBM patients, it is primarily informative in tumors with high MGMT pro-moter methylation. GBM patients with high MGMT promoter meth-ylation, who would be expected to respond well to alkylating agents, had similar survival to those with low MGMT promoter methylation if they also had high BCL3 expression. This finding is likely because cytotoxic O6-methylguanine adducts are required for BCL-3 to pro-mote resistance to TMZ. In the presence of high MGMT expression, cytotoxic adducts are repaired before they signal to BCL-3 (fig. S8).

B

C

EV

BCL-3

EV EVBCL-3BCL-3

sh-B

CL3

sh-S

CR

sh-S

CR

sh-B

CL3

A172 GBM43S GBM34 GBM44

E-CAD

VIM

BCL-3

SNAI1

TWIST1

GAPDH

2

4

6

8

10 r = 0.58, P < 0.0001

Nor

mal

ized

SN

AI1

m

RN

A

r = 0.58, P < 0.0001 r = 0.30, P < 0.0001

02468

1012

Nor

mal

ized

TW

IS

T1

mR

NA

10

12

14

16

18

20 r = 0.55, P < 0.0001

5 7 9 11 13Normalized BCL3

Nor

mal

ized

FN

1 m

RN

A

5 7 9 11 13Normalized BCL3

5 7 9 11 13Normalized BCL3

14

15

16

17

18

Nor

mal

ized

VIM

mR

NA

5 7 9 11 13Normalized BCL3

A

E

GSE4271

MES PN Pro

Nor

mal

ized

BC

L3

exp

ress

ion

5

8

6

7

10

9

TCGA GBM

MES PNNeural

Classical

Nor

mal

ized

BC

L3

exp

ress

ion

4

5

7

6

P = 0.0047P = 0.0015

P = 0.0179P < 0.0001

P < 0.0001

D

sh-SCRsh-BCL3 #1

0.0

0 5 10 15 20

0.5

1.0

1.5

2.0

2.5

0.0

EV

0 5 15 20 30

0.5

1.0

1.5

2.0

2.5

BCL-3

2510

sh-BCL3 #2

Dose (number of cells) Dose (number of cells)

Non

sphe

re fo

rmin

g (lo

g sc

ale)

Non

sphe

re fo

rmin

g (lo

g sc

ale)

*** **

GBM44 (BCL-3 low) GBM34 (BCL-3 high)

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

CDH1 FN1 VIM SNAI1 SNAI2 ZEB1 TWIST1

GBM43S sh-SCR

GBM43S sh-BCL3

Rel

ativ

e m

RN

A e

xpre

ssio

n

** ** **

**

**

A172 (BCL-3 low) GBM43S (BCL-3 high)High HighLow LowBCL-3:

Fig. 4. BCL-3 induces mesenchymal differentiation. (A) Photomicrograph of A172 cells stably expressing BCL-3 or EV (×20 magnification). (B) Quantitative PCR (left) and representative immunoblot (right) of mesenchymal markers in GSCs and GBM cells. qPCR data show mean value relative to GAPDH normalized to sh-SCR ± SD (n = 3). (C) Limiting dilution neurosphere assays in GBM44 and GBM34 GSCs expressing the indicated constructs (n = 2 biologic replicates). (D) Correlation between mesenchymal markers and BCL3 mRNA in TCGA GBM samples (n = 166 for all genes). (E) BCL3 mRNA expression in GBM molecular subtypes. Phillips (left) and Verhaak (right) classifica-tion schemes. For GSE4271: MES, n = 22; PN, n = 9; Proliferative (Pro), n = 24. For TCGA: MES, n = 53; Classical, n = 37; Neural, n = 24; PN, n = 53. *P < 0.05 and **P < 0.01. Pearson correlation analyzed by two-sided Student’s t test.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 9: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

9 of 15

A C D

I

J

2

CD44

CCL2

CXCL5

IER3

ICAM1

IRF1

IL1B

IL15

IL15RA

IL8

NQO1

PTGS2

PSMB9

S100A6

TFPI2

TNFAIP3

BCL2A1

CD48

CCR5

IL6

MMP9

PTX3

PLAU

SOD2

VCAM1

CSF2

IRF2

IRF7

IL11

NFKBIA

NOD2

STAT5A

HMOX1

CCL5

FAS

TNC

IL1A

VEGFC

si-Ctrl si-BCL3

Z score

2

5

8

11

14

Nor

mal

ized

IL8

mR

NA r = 0.59, P < 0.0001

r = 0.79, P < 0.0001

3

5

7

9

11

13

Nor

mai

lzed

LIF

mR

NA

8

10

12

14

16

Nor

mal

ized

CD

44

mR

NA

r = 0.63, P < 0.0001

4

6

8

10

12

14

Nor

mal

ized

CC

L2

mR

NA r = 0.68, P < 0.0001

5 7 9 11 135 7 9 11 13

5 7 9 11 13 5 7 9 11 13

GAPDH

CD44

BCL-3

CCL2G

BM

44 (B

CL-

3 lo

w)

BCL-3

EV

GAPDH

CD44

BCL-3

CCL2

GB

M34

(BC

L-3

high

)

sh-S

CR

sh-B

CL3

Lysate + + + + + +EV BCL-3

NF-κB

OCT1

LysateEV BCL-3

NF-κB

SS

+ + ++ ++ + + ++p50 p52 p65IgG p50 p52 p65Antibody

0

1

2

3

4

EV BCL-3

VehicleTNFα

Rela

tive

lucif

eras

e ac

tivity

**

**

Freeprobe

Freeprobe

Normalized BCL3 Normalized BCL3

Normalized BCL3 Normalized BCL3

IgG

LIF

Rel

ativ

e pr

omot

er e

nric

hmen

t

p50 p52 p65

CD44

Rel

ativ

e pr

omot

er e

nric

hmen

t

p50 p52 p65IP:

CCL2

Rel

ativ

e pr

omot

er e

nric

hmen

t

p50 p52 p65

*

*

**U87 EVU87 BCL-3

U87 EVU87 BCL-3

* **

U87 EVU87 BCL-3

**

**

SC SC NSNSCompetitor

IP: IP:

K

BCL-3

p65

Patient 1 Patient 2

BCL-3 low BCL-3 high

p65 lowp65 high

Fisher’s exact test, P = 0.0094

0.0

0.2

0.4

0.6

0.8

1.0

1.2

CD44 CCL2 IL1B IL15 IL8 LIF CSF2

GBM34 sh-SCR GBM34 sh-BCL3

Rel

ativ

e m

RN

A ex

pres

sion

** *** **** ****

B

BCL-3

p65

H3

A172

U25

1U

87T9

8G

BM26

GBM

34

GBM

44G

BM8

GBM

43S

SN28

1SN

186

G

H

0123456789

p65 p50 p52 IgG

GBM44GBM34

Rel

ativ

e pr

omot

er e

nric

hmen

t

10

12

0

2

4

6

8

p65 p50 p52 IgG

GBM44GBM34

Rel

ativ

e pr

omot

er E

nric

hmen

t

GBM44GBM34

0

1

2

3

4

p65 p50 p52 IgG

Rel

ativ

e pr

omot

er E

nric

hmen

tCD44 IL8 CCL2

GBM34 (BCL-3 high)

GBM44 (BCL-3 low) GBM34 (BCL-3 high)

U87 (BCL-3 intermediate)

E

p50

p65

p52

BCL-3

GAPDH

EV BCL-3Whole cell

p65

GAPDH

BCL-3

H3H3

p65

GAPDH

EV BCL-3EV BCL-3

p52 p52

p50 p50

Nuclear Cytoplasmic

p65

BCL-3

GAPDH

H3

p65

BCL-3

GAPDH

H3

sh-S

CR

sh-B

CL3 #1

F GBM34 (BCL-3 high)U87 (BCL-3 intermediate)

BCL-3

Nuclear Cytoplasmic

sh-B

CL3 #2

sh-S

CR

sh-B

CL3 #1

sh-B

CL3 #2

U87 (BCL-3 intermediate)

U87 (BCL-3 intermediate)U87 (BCL-3 intermed)

1 0 1 2

Fig. 5. BCL-3 promotes p65 nuclear translocation and NF-B dimer exchange. (A) Heatmap representing expression of all MES-specific NF-B target genes [identified in (10)] in microarray analysis of U87 cells expressing si-control (Ctrl) or si-BCL3, performed using three separate biological samples. Z score normalized expression is graded by color. (B) qPCR (upper) of MES-specific genes in GBM34 GSCs. Data show mean value relative to GAPDH normalized to sh-SCR ± SD (n = 3). Bottom: Representative immunoblots in GBM44 and GBM34 GSCs expressing the indicated construct. (C) Correlation between MES-specific genes and BCL3 mRNA in TCGA GBM samples (n = 166 for all genes). (D) Relative luciferase activity in U87 cells expressing BCL-3 or EV treated with TNF (5 ng/ml) or vehicle (12 hours). Data show mean value, normalized to vehicle-treated EV, ± SD (n = 4). (E) Representative immunoblot using the indicated cellular fractions from U87 cells expressing HA-BCL-3 or EV. (F) Immunoblot analysis with the indicated antibody using cytoplasmic and nuclear extract from GBM34 GSCs expressing two separate sh-BCL3 constructs or sh-SCR. Histone 3 (H3) antibody was used as nuclear loading control. (G) Representative immunoblot using nuclear fractions from the indicated GSCs and GBM cell lines. (H) Representative IHC images (left) of GBM samples from glioma TMAs showing nuclear p65 and BCL-3 staining in two patients. Table (right) shows numbers of tumors in each category. (I) ChIP qPCR in the indicated cells after IP. Data represent enrichment at the indicated promoter as a percentage of input ± SD (n = 2). (J) Representative electrophoretic mobility shift assay (EMSA) with CD44 B probe using nuclear extracts from U87 cells expressing BCL-3 or EV. Supershift (SS) (right) and competition with specific (SC) or nonspecific (NS) probe (left); OCT1 binding confirms equal loading. (K) ChIP qPCR in GSCs after IP with the indicated antibodies. Data represent enrichment at the indicated promoter as a percentage of input ± SD (n = 2). *P < 0.05 and **P < 0.01. Pearson correlation analyzed by two-sided Student’s t test. Significance in IHC was calculated using two-sided Fisher’s exact test.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 10: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

10 of 15

B

I

A

M

H

Genes up-regulated by TMZ in si-Ctrl not si-BCL3

Genes down-regulated with si-BCL3

CDK5RAP2

FAS

FST

GPER

IDH2

MYLIP

RTTN

TERF1

TMEM194A

TOP2A

CD44

CCL2

IL1B

IL15

IL8

PTGS2

PSMB9

CSF2

....

10 13181

CAII

Rel

ativ

e C

AII e

xpre

ssio

n

0.0

1.0

2.0

3.0

4.0

24 48 72

DMSOTMZ 100 µM

**

**

*

IP: p52

Rel

ativ

e C

hrom

atin

enr

ichm

ent

0

2

4

6

8

10

12 DMSOTMZ**

**

κB1 κB2

SC SC+ + + + + +DMSO TMZ

CompetitorLysate

NF-κB

DMSO TMZ

IgG+ + ++ ++ + + ++p50 p52 p65 p50 p52 p65Antibody

Lysate

SS

NF-κB

Freeprobe

Freeprobe

OCT1

D

P = 0.0081

Nor

mal

ized

CA

II m

RN

A

8

6

12

Low HighBCL3

REMBRANDT GBM

10

P = 0.0219GSE4271

Nor

mal

ized

CA

II m

RN

A

8

6

10

12

Low HighBCL3Time (h)

01234567

κB1

DMSOTMZ

Rel

ativ

e pr

omot

er e

nric

hmen

t

****

κB2

IP: p50

IgG

si-Ctrl si-CAII

CAIIGAPDH

si-CAII

si-Ctrl

Rel

ativ

e ce

ll de

ath

0

1

2

3

4

5

6*

*

*

- 50 100 200TMZ (µM)

NSNS

C

Inpu

tIP

: BC

L-3

IP: F

LAG

IB: FLAGTMZ + + +

EV WT S329A

IB: BCL-3TMZ + + +

EV WT S329A

IB: BCL-3

IB: FLAGTMZ + + +

EV WT S329A

p50

IB: p50

IB: H3

IB: p52

IB: BCL-3

IB: p52

IB: p50

Inpu

t

TMZGBM34 GBM43S

IP BCL-3 IgG BCL-3 IgG

IgG heavy chain

J K

GGGATGTCCCGGGCATTTTCGGGGCTTCCC

κB1:κB2:κB3:

κB1 κB2 κB3Exon 1 Exon 2

+398 +714 +903+1

CAII

GBM26 (BCL-3 high)L

Rel

ativ

e ce

ll de

ath

CAIIGAPDH

CAIIEV

0.0

1.0

2.0

3.0

4.0

5.0

6.0

7.0

EVCAII**

**

- 100 200TMZ (µM)

GBM44 (BCL-3 low)

G

GBM43S (BCL-3 high)

^^^ ^

^

^^

^

^

U87 (BCL-3 intermediate)

Rel

ativ

e C

AII e

xpre

ssio

n **

GBM34 (BCL-3 high)

SCR BCL3

#1

DMSO

TMZ 100 µM

BCL3

#2sh:

E

0.0

0.5

1.0

1.5

2.0

2.5

3.0

3.5

4.0

CAII BCL3 MGMT

si-Ctrl/DMSO si-Ctrl/TMZ si-MGMT/DMSO si-MGMT/TMZ

Rel

ativ

e m

RN

A e

xpre

ssio

n

**F

0.0

0.5

1.0

1.5

2.0

2.5

3.0

SCRsh- p52/NFKB2

#2

p52/NFKB2

#1

DMSOTMZ 100 µM

GBM34 (BCL-3 high)R

elat

ive

CA

II ex

pres

sion

sh-p

52 #

2

GAPDH

p52sh

-SC

Rsh

-p52

#1

GBM26 (BCL-3 high/ MGMT high)

N

Sur

vivi

ng fr

actio

n

0.0

0.2

0.4

0.6

0.8

1.0CAIIGAPDH

si-CAII

si-Ctrl

****

****

- 25 50TMZ (µM)

si-Ctrl si-BCL3 si-BCL3 + si-CAII

U87 (BCL-3 intermediate)

**

(BCL-3 high)

U87 (BCL-3 intermediate)

U87 (BCL-3 intermediate)

Fig. 6. CAII mediates BCL-3–dependent resistance to TMZ. (A) Significantly (P < 0.05) altered transcripts in U87 cells expressing si-BCL3 or si-control treated with TMZ (100 M) or vehicle (24 hours). (B) CAII mRNA expression relative to GAPDH in GBM43S GSCs treated with TMZ (n = 3). (C) CAII mRNA in GBM34 GSCs expressing sh-BCL3 constructs or sh-control treated with vehicle or TMZ (48 hours) (n = 3). (D) CAII mRNA expression in patients from the indicated data set with lower and upper 33% BCL3 expression. REMBRANDT, n = 61 per group; GSE4271, n = 20 per group. (E) Expression of CAII, BCL3, and MGMT mRNA in GBM26 GSCs that express high MGMT (see fig. S7K) transfected with si-MGMT or si-control after treatment with 100 M TMZ for 48 hours. Data show mean expression relative to GAPDH normalized to si-control, dimethyl sulfoxide (DMSO)–treated, ± SD (n = 2). (F) CAII expression relative to GAPDH in GBM34 GSCs expressing sh-control or two separate sh-p52/NFKB2 constructs after treat-ment with TMZ (48 hours). Data show mean value relative to GAPDH ± SD (n = 3). Inset: Immunoblot with anti-p52. (G) Location and sequence of putative human CAII B sites. (H) ChIP qPCR in U87 cells treated with 100 M TMZ or vehicle (24 hours). Data are normalized to immunoglobulin G (IgG) and represent enrichment as a percentage of input ± SD (n = 2). (I) Representative EMSA with B2 probe in U87 nuclear lysate treated as in (H). Supershift (SS) (right) and competition with specific (SC) or nonspecific (NS) probe (left). OCT1 binding confirms equal loading. (J) Co-IP in GBM34 and GBM43S GSCs (both BCL-3 high) after treatment with TMZ (100 M, 24 hours). IP and IB were performed with the indicated antibodies. Arrowheads indicate IgG heavy chain band (n = 2). (K) Reciprocal co-IP in 293T cells expressing FLAG-p50-wt, FLAG-p50-S329A, or EV after treatment with vehicle or TMZ (100 M, 24 hours) (n = 2). (L and M) Trypan blue assays in GBM44 GSCs expressing HA-CAII or EV (L) and GBM26 GSCs expressing si-CAII or si-control (M) treated with TMZ (72 hours). Data show fold change in percent dead cells relative to vehicle ± SD (n = 3). Insets: Immunoblots with anti-CAII antibody. (N) Clonogenic assay in U87 cells expressing si-control or si-BCL3 with and without si-CAII treated with TMZ (n = 3). *P < 0.05 and **P < 0.01, two-sided Student’s t test.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 11: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

11 of 15

BCL-3 was previously found to be elevated in gliomas and related to survival (42); however, in that study, patients with all grades of glioma were analyzed together, precluding determination of prognostic value. Our results indicate that only tumors with low BCL3 expression will likely benefit from adding TMZ to IR, an important clinical obser-vation given that TMZ induces deleterious hypermutation that can cause malignant progression (43).

The data demonstrate that, in glioma, BCL3 expression is regulated by genetic, and epigenetic, modifications and that these alterations are linked to patient outcome. We see that BCL3 CN loss occurs as a result of modifications targeted to the chromosomal band 19q13. This finding, when considered with the observation that BCL-3 is a candidate onco-protein that has never been identified as a glioma driver (4), indicates that BCL3 loss is a passenger event unrelated to glioma formation. Although

the ability of passenger events to promote unintended therapeutic sus-ceptibility has been shown in animal models (44, 45), the link between loss of BCL3 and TMZ susceptibility demonstrates the importance of passenger modification to chemosensitivity in a clinical setting. The ef-fect of BCL3 deletion in glioma is particularly relevant given that alter-ations of 19q play an important role in modulating patient outcome in these tumors. Specifically, 19q, with 1p, co-deletion is predictive of re-sponse to alkylating chemotherapy in oligodendroglioma (46), whereas loss of 19q13 alone is associated with long-term survival in GBM (29, 47). The most widely accepted hypothesis as to why 19q13 loss is so preva-lent in gliomas is that a glioma- specific tumor suppressor is present in the region (48). Although no 19q tumor suppressor has yet been iden-tified in astrocytic tumors or GBM, capicua transcriptional repressor (CIC) was recently discovered in oligodendroglioma (49).

B C D

F

A

TMZ ++ ++ACZ ++

0.0

0.2

0.4

0.6

0.8

1.0

1.2

Rel

ativ

e su

rviv

ing

fract

ion si-CAIIsi-Ctrl

*CAII

GAPDH

si-C

trlsi

- CA

II

si-C

trlsi

- CA

II

U251U87

IB: CAIIIB: GAPDH

si-C

trlsi

- CA

II

si-C

trlsi

-CA

II

mR

NA

Pro

tein

U251U87

0.0

0.2

0.4

0.6

0.8

1.0

Sur

viva

l fac

tor

15 30 45Days post-graft

60 75 90

VehicleTMZ

TMZ + ACZACZ

H

0.0

0.2

0.4

0.6

0.8

1.0

Sur

viva

l fac

tor

Days post-graft

VehicleACZTMZTMZ + ACZ

0 5 10 15 20 25 30 35

I sh-SCR/vehiclesh-SCR/TMZ

sh-BCL3/Vehiclesh-BCL3/ACZsh-BCL3/TMZsh-BCL3/ACZ + TMZ

0.0

0.2

0.4

0.6

0.8

1.0

Sur

viva

l fac

tor

0 10 20 30 40 50 60Days post-graft

GAPDH

BCL-3

sh-S

CR

sh- B

CL

3

0 10 20 30 40 50 60 70 80

VehicleACZTMZTMZ + ACZ

Days post-graft

0.0

0.2

0.4

0.6

0.8

1.0

Surv

ival

fact

orG

0

2

4

6

8sh-SCRsh-SCR + ACZsh-BCL3

sh-BCL3 + ACZ

- 50 100TMZ (µM)

GBM34 (BCL-3 high)

Rel

ativ

e ce

ll de

ath

**

**

0

2

4

6

8

- 100 200TMZ (µM)

Rel

ativ

e ce

ll de

ath

EVEV + ACZBCL-3BCL-3 + ACZ **

**

GBM44 (BCL-3 low)

U87 (BCL-3 intermediate) GBM43S (BCL-3 high)

GBM34 (BCL-3 high) GBM34 (BCL-3 high)

1 2 3++

++

0.0

0.2

0.4

0.6

0.8

1.0

1.2

Sur

vivi

ng fr

actio

n

TMZACZ

U251

E

CAII (HA)

GAPDH

CAIIEV

*

*

0.0

0.2

0.4

0.6

0.8

1.0

1.2

Sur

vivi

ng fr

actio

n

DMSO ACZ

- 50 100TMZ (µM)

U87 (BCL-3 intermed)

U251 (BCL-3 intermediate)

***

*

Fig. 7. ACZ chemosensitizes GBM xenografts to TMZ. (A and B) Trypan blue assays at 72 hours in (A) GBM34 GSCs expressing sh-BCL3 or sh-SCR or (B) GBM44 GSCs expressing BCL-3 or EV treated with TMZ and/or 100 M ACZ as indicated. Data show fold change in percent dead cells relative to cells transfected with EV, or sh-SCR, treated with vehicle, ± SD (n = 3). (C) Clonogenic assay in U87 cells expressing si-CAII or si-control treated with 50 M TMZ with or without 100 M ACZ. Data show surviv-ing fraction relative to TMZ alone ± SD (n = 3). (D) PCR analysis of CAII mRNA and immunoblot analysis of CAII protein in U87 and U251 cells expressing si-control or si-CAII. Clonogenic assay (right) in U251 cells treated with 100 M TMZ and/or 100 M ACZ (n = 3). (E) Clonogenic assay in U251 cells stably expressing CAII or EV treated with TMZ and/or 100 M ACZ (n = 3). (F) Kaplan-Meier curves of mice bearing intracranial U87 tumors (n = 5 per group) treated with TMZ on days 5, 7, and 9 (5 mg/kg per dose) and/or ACZ on days 5 to 26 (15 mg/kg per day). (G) Kaplan-Meier curves of mice bearing intracranial GBM43S PDX (n = 7 per group) treated with TMZ on days 5, 7, and 9 (5 mg/kg per dose) and/or ACZ on days 5 to 26 (15 mg/kg per day). (H) Kaplan-Meier curves of mice bearing intracranial GBM34 PDX (n = 7 per group) treated with TMZ on days 5, 7, and 9 (10 mg/kg per dose) and/or ACZ on days 5 to 26 (15 mg/kg per day). (I) Kaplan-Meier curves of mice bearing intracranial GBM34 PDX tumors stably expressing either sh-BCL3 (n = 6 per group) or sh-SCR (n = 5 per group) treated with TMZ on days 5, 7, and 9 (10 mg/kg per dose) and/or ACZ on days 5 to 26 (15 mg/kg per day). Inset: Immuno blot of cells from the indicated tumors. *P < 0.05 and **P < 0.02.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 12: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

12 of 15

Our studies indicate that BCL-3 promotes GBM mesenchymal dif-ferentiation, an observation consistent with previous reports linking BCL-3 to EMT (50–52). We find that, in GBM cells, BCL-3 up-regulated EMT markers and that, in clinical GBM, BCL3 expression correlated strongly with MES signature gene expression. From a mechanistic standpoint, high BCL-3 led to a change in the composition of the NF-B dimer at MES gene promoters involving replacement of p50 by p52. In addition, BCL-3 induced p65 nuclear translocation, a finding consistent with the known importance of this subunit in MES change (10). Whereas previous work demonstrates a cell-extrinsic pathway for activation of p65 in GBM cells by cytokines released from infiltrating macrophages and microglia (10), genetic and epigenetic regulation of BCL3 represents mechanisms by which cell-intrinsic pathways also con-tribute to promoting NF-B–dependent mesenchymal differentiation.

An important feature of predictors like BCL-3 is that they are also informative in that they can identify pathways to improve treatment response. MGMT promoter methylation is one such predictor; how-ever, inhibiting MGMT has not proven to be an effective chemosen-sitizing strategy clinically (8). Inhibition of BCL-3 is not currently feasible in patients; therefore, we searched for downstream BCL-3– regulated targets and identified CAII as a mediator of BCL-3–dependent resistance to TMZ. Although several factors have previously been re-ported to contribute to the antiapoptotic effects of BCL-3 (53–55), from a clinical perspective, CAII stands out because it can be effec-tively inhibited by ACZ. CAII mRNA expression is not prognostic in untreated GBM (GlioVis data portal) (32), yet elevated endothelial CAII has been associated with worse outcome in astrocytoma (56). Regardless of these observations, our data demonstrate that it is not basal expression but the induction of CAII by TMZ that is important in modulating response to therapy.

Although we demonstrated the predictive value of BCL3 in several independent data sets, an important limitation of our study is its ret-rospective nature. Ultimately, the clinical validity of using BCL-3 as a predictor in GBM will require verification in a prospective randomized trial. In addition, the question of which CA inhibitor is best for clinical use with TMZ will require further analysis. Finally, a mechanistic limita-tion of our study is that we have yet to determine how BCL-3 promotes p65 nuclear translocation and promoter-specific dimer exchange.

ACZ has not previously been examined in GBM xenografts; how-ever, it was initially shown to promote chemosensitization in murine fibrosarcoma (57). More recently, ACZ was found to promote cyto-toxicity of GBM cells in vitro and other cancers in vivo (37–39, 58). Although there has been significant interest in targeting other hypoxia- specific CA isoforms for GBM therapy (41, 59), ACZ is attractive be-cause of its general clinical use and well-tolerated dosing profile (40). Using PDXs, we see that, in GBM, ACZ promoted chemosensitization specifically in the presence of high BCL-3 and low MGMT expression. Our studies suggest that, in the clinical setting, repurposing ACZ might be particularly effective in a subgroup of MGMT promoter methylated tumors that have high BCL-3 expression. Given the increase in mo-lecular analysis of gliomas, factors such as BCL-3 might assume an important role in individualizing patient treatment, a strategy occur-ring with increasing frequency in cancer therapy.

MATERIALS AND METHODSStudy designThe objectives of this study were to examine the role of BCL-3 in the response of glioma to alkylating chemotherapy and to evaluate the

use of ACZ as a chemosensitizer in experimental GBM. There are several design aspects relevant in this work. First, to examine the pre-dictive role of BCL3, initially, TCGA was examined and the results were subsequently validated in other databases. Numbers of patients included or excluded in each data set are specifically noted in Materials and Methods and in each individual figure panel. To investigate sur-vival based on BCL-3 IHC, we performed an initial power analysis based on the mRNA data from TCGA. This gave n = 74 and 32 patients per group, respectively, to have 80% power to detect a significant difference between low and high BCL-3 expression at a two-sided P < 0.05. Subsequently, after institutional review board approval, we obtained 86 consecutive GBM samples that had adequate tissue for IHC. Additional patients were not recruited. Patients were excluded from survival analysis if they died before treatment due to infection or massive hemorrhage, if no follow-up was available, or if they initially had an LGG that progressed to GBM. IHC and FISH grading was performed by two independent investigators blinded to diagnosis and to survival.

For animal studies, no statistical method was used to determine sample size. Efforts were made to achieve the scientific goals with the minimum number of animals. A sample size of five to seven ani-mals per group was chosen on the basis of our previous experience using intracranial PDX GBMs, where we have observed 100% tumor engraftment success. After tumor implantation and before treatment, animals were randomized into the different treatment groups. Animals were excluded from the study if they were sacrificed before treatment/randomization. Determination of the survival time was performed blinded to the specific treatment group. Experiments were repeated by more than one individual to ensure reproducibility. The survival end-point was reached when mice lost at least 20% body weight or showed symptoms of neurological deficit. Raw data are located in table S17.

Statistical analysisData analysis was performed using Stata/IC 13.0 statistical software (Stata Corporation, licensed to the University of Chicago, Research Computing Center). The Cox proportional hazard model was used for both univariate and multivariate analyses using the specific co-variates noted in Results. For survival studies, Kaplan-Meier curves were plotted, and the log-rank test was performed for comparison of cohorts. HR and 95% CIs were calculated using the Mantel- Haenszel estimator model. For analysis of 19q CN correlation, correlation matrix analysis was used, and the R/corrplot package was used to display the correlation matrix. For box-and-whisker graphs, boxes show median and 25th and 75th percentiles, whereas whiskers show the 5th and 95th percentiles analyzed by unpaired t test. In vitro and other studies as indicated were analyzed by two-tailed Student’s t test with significance taken as P < 0.05. Pearson correlation was also ana-lyzed by two-tailed Student’s t test.

SUPPLEMENTARY MATERIALSwww.sciencetranslationalmedicine.org/cgi/content/full/10/448/eaar2238/DC1MethodsFig. S1. BCL3 predicts response to alkylating chemotherapy in GBM.Fig. S2. BCL3 is relevant in LGGs and pan-glioma patients.Fig. S3. Loss of BCL3 is a passenger event associated with survival.Fig. S4. BCL-3 induces mesenchymal differentiation.Fig. S5. BCL-3 promotes p65 nuclear translocation and NF-B dimer exchange.Fig. S6. CAII mediates BCL-3–dependent resistance to TMZ.Fig. S7. ACZ chemosensitizes GBM xenografts to TMZ.Fig. S8. Model demonstrating role of BCL-3 in promoting resistance to TMZ.Table S1. Molecular profile of GBM cell lines.

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 13: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

13 of 15

Table S2. Cox regression analysis using different cutoff points for BCL3 expression in TCGA GBM patients.Table S3. Multivariate Cox regression analysis in TCGA GBM patients treated with and without alkylating chemotherapy (TMZ).Table S4. Clinical features of University of Chicago GBM patients.Table S5. Cox regression analysis using different BCL3 expression cutoff points in TCGA LGG patients.Table S6. Cox regression analysis in TCGA LGG patients with and without alkylating chemotherapy.Table S7. Multivariate Cox regression analysis in patients from EORTC 26951 treated either with or without alkylating chemotherapy.Table S8. Cox regression analysis of TCGA pan-glioma patients who received alkylating chemotherapy.Table S9. Multivariate Cox regression of TCGA pan-glioma patients incorporating BCL3 CN.Table S10. Multivariate Cox regression of TCGA pan-glioma patients incorporating BCL3 promoter methylation.Table S11. Correlation between BCL3 mRNA expression and the expression of each MES or PN signature gene in TCGA GBM patients.Table S12. Genes that are up- and down-regulated (at least 1.5-fold) in U87 cells expressing si-BCL3 relative to those expressing si-control (see separate Excel file).Table S13. Genes that are significantly (adjusted P < 0.05) up- and down-regulated in U87 cells expressing si-BCL3 or si-control after treatment with TMZ compared to vehicle (see separate Excel file).Table S14. Oligonucleotides (oligos) for EMSA.Table S15. ChIP primers and target gene B sites.Table S16. Oligonucleotide sequences for qPCR.Table S17. Raw data (see separate Excel file).References (60–72)

REFERENCES AND NOTES 1. H. S. Phillips, S. Kharbanda, R. Chen, W. F. Forrest, R. H. Soriano, T. D. Wu, A. Misra,

J. M. Nigro, H. Colman, L. Soroceanu, P. M. Williams, Z. Modrusan, B. G. Feuerstein, K. Aldape, Molecular subclasses of high-grade glioma predict prognosis, delineate a pattern of disease progression, and resemble stages in neurogenesis. Cancer Cell 9, 157–173 (2006).

2. R. G. W. Verhaak, K. A. Hoadley, E. Purdom, V. Wang, Y. Qi, M. D. Wilkerson, C. R. Miller, L. Ding, T. Golub, J. P. Mesirov, G. Alexe, M. Lawrence, M. O’Kelly, P. Tamayo, B. A. Weir, S. Gabriel, W. Winckler, S. Gupta, L. Jakkula, H. S. Feiler, J. G. Hodgson, C. D. James, J. N. Sarkaria, C. Brennan, A. Kahn, P. T. Spellman, R. K. Wilson, T. P. Speed, J. W. Gray, M. Meyerson, G. Getz, C. M. Perou, D. N. Hayes; Cancer Genome Atlas Research Network, Integrated genomic analysis identifies clinically relevant subtypes of glioblastoma characterized by abnormalities in PDGFRA, IDH1, EGFR, and NF1. Cancer Cell 17, 98–110 (2010).

3. R. Stupp, W. P. Mason, M. J. van den Bent, M. Weller, B. Fisher, M. J. B. Taphoorn, K. Belanger, A. A. Brandes, C. Marosi, U. Bogdahn, J. Curschmann, R. C. Janzer, S. K. Ludwin, T. Gorlia, A. Allgeier, D. Lacombe, J. G. Cairncross, E. Eisenhauer, R. O. Mirimanoff; European Organisation for Research and Treatment of Cancer Brain Tumor and Radiotherapy Groups; National Cancer Institute of Canada Clinical Trials Group, Radiotherapy plus concomitant and adjuvant temozolomide for glioblastoma. N. Engl. J. Med. 352, 987–996 (2005).

4. C. W. Brennan, R. G. W. Verhaak, A. McKenna, B. Campos, H. Noushmehr, S. R. Salama, S. Zheng, D. Chakravarty, J. Z. Sanborn, S. H. Berman, R. Beroukhim, B. Bernard, C.-J. Wu, G. Genovese, I. Shmulevich, J. Barnholtz-Sloan, L. Zou, R. Vegesna, S. A. Shukla, G. Ciriello, W. K. Yung, W. Zhang, C. Sougnez, T. Mikkelsen, K. Aldape, D. D. Bigner, E. G. Van Meir, M. Prados, A. Sloan, K. L. Black, J. Eschbacher, G. Finocchiaro, W. Friedman, D. W. Andrews, A. Guha, M. Iacocca, B. P. O’Neill, G. Foltz, J. Myers, D. J. Weisenberger, R. Penny, R. Kucherlapati, C. M. Perou, D. N. Hayes, R. Gibbs, M. Marra, G. B. Mills, E. Lander, P. Spellman, R. Wilson, C. Sander, J. Weinstein, M. Meyerson, S. Gabriel, P. W. Laird, D. Haussler, G. Getz, L. Chin; TCGA Research Network, The somatic genomic landscape of glioblastoma. Cell 155, 462–477 (2013).

5. B. G. Masayesva, P. Ha, E. Garrett-Mayer, T. Pilkington, R. Mao, J. Pevsner, T. Speed, N. Benoit, C.-S. Moon, D. Sidransky, W. H. Westra, J. Califano, Gene expression alterations over large chromosomal regions in cancers include multiple genes unrelated to malignant progression. Proc. Natl. Acad. Sci. U.S.A. 101, 8715–8720 (2004).

6. E. Frei III, Gene deletion: A new target for cancer chemotherapy. Lancet 342, 662–664 (1993).

7. S. Tanaka, D. N. Louis, W. T. Curry, T. T. Batchelor, J. Dietrich, Diagnostic and therapeutic avenues for glioblastoma: No longer a dead end? Nat. Rev. Clin. Oncol. 10, 14–26 (2013).

8. M. E. Hegi, L. Liu, J. G. Herman, R. Stupp, W. Wick, M. Weller, M. P. Mehta, M. R. Gilbert, Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylation with

clinical outcomes in glioblastoma and clinical strategies to modulate MGMT activity. J. Clin. Oncol. 26, 4189–4199 (2008).

9. A. Murat, E. Migliavacca, T. Gorlia, W. L. Lambiv, T. Shay, M.-F. Hamou, N. de Tribolet, L. Regli, W. Wick, M. C. M. Kouwenhoven, J. A. Hainfellner, F. L. Heppner, P.-Y. Dietrich, Y. Zimmer, J. G. Cairncross, R.-C. Janzer, E. Domany, M. Delorenzi, R. Stupp, M. E. Hegi, Stem cell–related “self-renewal” signature and high epidermal growth factor receptor expression associated with resistance to concomitant chemoradiotherapy in glioblastoma. J. Clin. Oncol. 26, 3015–3024 (2008).

10. K. P. L. Bhat, V. Balasubramaniyan, B. Vaillant, R. Ezhilarasan, K. Hummelink, F. Hollingsworth, K. Wani, L. Heathcock, J. D. James, L. D. Goodman, S. Conroy, L. Long, N. Lelic, S. Wang, J. Gumin, D. Raj, Y. Kodama, A. Raghunathan, A. Olar, K. Joshi, C. E. Pelloski, A. Heimberger, S. H. Kim, D. P. Cahill, G. Rao, W. F. A. Den Dunnen, H. W. G. M. Boddeke, H. S. Phillips, I. Nakano, F. F. Lang, H. Colman, E. P. Sulman, K. Aldape, Mesenchymal differentiation mediated by NF-B promotes radiation resistance in glioblastoma. Cancer Cell 24, 331–346 (2013).

11. A. M. Schmitt, C. D. Crawley, S. Kang, D. R. Raleigh, X. Yu, J. S. Wahlstrom, D. J. Voce, T. E. Darga, R. R. Weichselbaum, B. Yamini, p50 (NF-B1) is an effector protein in the cytotoxic response to DNA methylation damage. Mol. Cell 44, 785–796 (2011).

12. K. E. Cahill, R. A. Morshed, B. Yamini, Nuclear factor-B in glioblastoma: Insights into regulators and targeted therapy. Neuro Oncol. 18, 329–339 (2016).

13. S. Palmer, Y. H. Chen, Bcl-3, a multifaceted modulator of NF-B-mediated gene transcription. Immunol. Res. 42, 210–218 (2008).

14. P. C. Cogswell, D. C. Guttridge, W. K. Funkhouser, A. S. Baldwin Jr., Selective activation of NF-kappa B subunits in human breast cancer: Potential roles for NF-kappa B2/p52 and for Bcl-3. Oncogene 19, 1123–1131 (2000).

15. N. J. Thornburg, R. Pathmanathan, N. Raab-Traub, Activation of nuclear factor-B p50 homodimer/Bcl-3 complexes in nasopharyngeal carcinoma. Cancer Res. 63, 8293–8301 (2003).

16. T. Fujita, G. P. Nolan, H. C. Liou, M. L. Scott, D. Baltimore, The candidate proto-oncogene bcl-3 encodes a transcriptional coactivator that activates through NF-kappa B p50 homodimers. Genes Dev. 7, 1354–1363 (1993).

17. V. Bours, G. Franzoso, V. Azarenko, S. Park, T. Kanno, K. Brown, U. Siebenlist, The oncoprotein Bcl-3 directly transactivates through kappa B motifs via association with DNA-binding p50B homodimers. Cell 72, 729–739 (1993).

18. V. Y.-F. Wang, Y. Li, D. Kim, X. Zhong, Q. Du, M. Ghassemian, G. Ghosh, Bcl3 phosphorylation by Akt, Erk2, and IKK is required for its transcriptional activity. Mol. Cell 67, 484–497.e5 (2017).

19. M. Bredel, D. M. Scholtens, A. K. Yadav, A. A. Alvarez, J. J. Renfrow, J. P. Chandler, I. L. Y. Yu, M. S. Carro, F. Dai, M. J. Tagge, R. Ferrarese, C. Bredel, H. S. Phillips, P. J. Lukac, P. A. Robe, A. Weyerbrock, H. Vogel, S. Dubner, B. Mobley, X. He, A. C. Scheck, B. I. Sikic, K. D. Aldape, A. Chakravarti, G. R. Harsh IV, NFKBIA deletion in glioblastomas. N. Engl. J. Med. 364, 627–637 (2011).

20. S. Madhavan, J.-C. Zenklusen, Y. Kotliarov, H. Sahni, H. A. Fine, K. Buetow, Rembrandt: Helping personalized medicine become a reality through integrative translational research. Mol. Cancer Res. 7, 157–167 (2009).

21. P. Bady, D. Sciuscio, A.-C. Diserens, J. Bloch, M. J. van den Bent, C. Marosi, P.-Y. Dietrich, M. Weller, L. Mariani, F. L. Heppner, D. R. McDonald, D. Lacombe, R. Stupp, M. Delorenzi, M. E. Hegi, MGMT methylation analysis of glioblastoma on the Infinium methylation BeadChip identifies two distinct CpG regions associated with gene silencing and outcome, yielding a prediction model for comparisons across datasets, tumor grades, and CIMP-status. Acta Neuropathol. 124, 547–560 (2012).

22. L. Erdem-Eraslan, L. A. Gravendeel, J. de Rooi, P. H. C. Eilers, A. Idbaih, W. G. M. Spliet, W. F. A. den Dunnen, J. L. Teepen, P. Wesseling, P. A. E. Sillevis Smitt, J. M. Kros, T. Gorlia, M. J. van den Bent, P. J. French, Intrinsic molecular subtypes of glioma are prognostic and predict benefit from adjuvant procarbazine, lomustine, and vincristine chemotherapy in combination with other prognostic factors in anaplastic oligodendroglial brain tumors: A report from EORTC study 26951. J. Clin. Oncol. 31, 328–336 (2013).

23. D. N. Louis, A. Perry, G. Reifenberger, A. von Deimling, D. Figarella-Branger, W. K. Cavenee, H. Ohgaki, O. D. Wiestler, P. Kleihues, D. W. Ellison, The 2016 World Health Organization Classification of Tumors of the Central Nervous System: A summary. Acta Neuropathol. 131, 803–820 (2016).

24. M. Ceccarelli, F. P. Barthel, T. M. Malta, T. S. Sabedot, S. R. Salama, B. A. Murray, O. Morozova, Y. Newton, A. Radenbaugh, S. M. Pagnotta, S. Anjum, J. Wang, G. Manyam, P. Zoppoli, S. Ling, A. A. Rao, M. Grifford, A. D. Cherniack, H. Zhang, L. Poisson, C. G. Carlotti Jr., D. P. da Cunha , A. Rao, T. Mikkelsen, C. C. Lau, W. K. A. Yung, R. Rabadan, J. Huse, D. J. Brat, N. L. Lehman, J. S. Barnholtz-Sloan, S. Zheng, K. Hess, G. Rao, M. Meyerson, R. Beroukhim, L. Cooper, R. Akbani, M. Wrensch, D. Haussler, K. D. Aldape, P. W. Laird, D. H. Gutmann; TCGA Research Network, H. Noushmehr, A. Iavarone, R. G. W. Verhaak, Molecular profiling reveals biologically discrete subsets and pathways of progression in diffuse glioma. Cell 164, 550–563 (2016).

25. A. von Deimling, B. Bender, R. Jahnke, A. Waha, J. Kraus, S. Albrecht, R. Wellenreuther, F. Faßbender, J. Nagel, A. G. Menon, D. N. Louis, D. Lenartz, J. Schramm, O. D. Wiestler,

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 14: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

14 of 15

Loci associated with malignant progression in astrocytomas: A candidate on chromosome 19q. Cancer Res. 54, 1397–1401 (1994).

26. J. S. Smith, B. Alderete, Y. Minn, T. J. Borell, A. Perry, G. Mohapatra, S. M. Hosek, D. Kimmel, J. O’Fallon, A. Yates, B. G. Feuerstein, P. C. Burger, B. W. Scheithauer, R. B. Jenkins, Localization of common deletion regions on 1p and 19q in human gliomas and their association with histological subtype. Oncogene 18, 4144–4152 (1999).

27. M. Nakamura, F. Yang, H. Fujisawa, Y. Yonekawa, P. Kleihues, H. Ohgaki, Loss of heterozygosity on chromosome 19 in secondary glioblastomas. J. Neuropathol. Exp. Neurol. 59, 539–543 (2000).

28. M. Weller, J. Felsberg, C. Hartmann, H. Berger, J. P. Steinbach, J. Schramm, M. Westphal, G. Schackert, M. Simon, J. C. Tonn, O. Heese, D. Krex, G. Nikkhah, T. Pietsch, O. Wiestler, G. Reifenberger, A. von Deimling, M. Loeffler, Molecular predictors of progression-free and overall survival in patients with newly diagnosed glioblastoma: A prospective translational study of the German Glioma Network. J. Clin. Oncol. 27, 5743–5750 (2009).

29. E. C. Burton, K. R. Lamborn, B. G. Feuerstein, M. Prados, J. Scott, P. Forsyth, S. Passe, R. B. Jenkins, K. D. Aldape, Genetic aberrations defined by comparative genomic hybridization distinguish long-term from typical survivors of glioblastoma. Cancer Res. 62, 6205–6210 (2002).

30. R. Beroukhim, G. Getz, L. Nghiemphu, J. Barretina, T. Hsueh, D. Linhart, I. Vivanco, J. C. Lee, J. H. Huang, S. Alexander, J. Du, T. Kau, R. K. Thomas, K. Shah, H. Soto, S. Perner, J. Prensner, R. M. Debiasi, F. Demichelis, C. Hatton, M. A. Rubin, L. A. Garraway, S. F. Nelson, L. Liau, P. S. Mischel, T. F. Cloughesy, M. Meyerson, T. A. Golub, E. S. Lander, I. K. Mellinghoff, W. R. Sellers, Assessing the significance of chromosomal aberrations in cancer: Methodology and application to glioma. Proc. Natl. Acad. Sci. U.S.A. 104, 20007–20012 (2007).

31. D. W. Lee, D. Ramakrishnan, J. Valenta, I. F. Parney, K. J. Bayless, R. Sitcheran, The NF-B RelB protein is an oncogenic driver of mesenchymal glioma. PLOS ONE 8, e57489 (2013).

32. R. L. Bowman, Q. Wang, A. Carro, R. G. W. Verhaak, M. Squatrito, GlioVis data portal for visualization and analysis of brain tumor expression datasets. Neuro Oncol. 19, 139–141 (2017).

33. S. A. Mani, W. Guo, M.-J. Liao, E. Ng. Eaton, A. Ayyanan, A. Y. Zhou, M. Brooks, F. Reinhard, C. C. Zhang, M. Shipitsin, L. L. Campbell, K. Polyak, C. Brisken, J. Yang, R. A. Weinberg, The epithelial-mesenchymal transition generates cells with properties of stem cells. Cell 133, 704–715 (2008).

34. B. Manavalan, S. Basith, Y.-M. Choi, G. Lee, S. Choi, Structure-function relationship of cytoplasmic and nuclear IB proteins: An in silico analysis. PLOS ONE 5, e15782 (2010).

35. B. Masereel, S. Rolin, F. Abbate, A. Scozzafava, C. T. Supuran, Carbonic anhydrase inhibitors: Anticonvulsant sulfonamides incorporating valproyl and other lipophilic moieties. J. Med. Chem. 45, 312–320 (2002).

36. H. M. Said, C. T. Supuran, C. Hageman, A. Staab, B. Polat, A. Katzer, A. Scozzafava, J. Anacker, M. Flentje, D. Vordermark, Modulation of carbonic anhydrase 9 (CA9) in human brain cancer. Curr. Pharm. Des. 16, 3288–3299 (2010).

37. H. M. Said, C. Hagemann, F. Carta, A. Katzer, B. Polat, A. Staab, A. Scozzafava, J. Anacker, G. H. Vince, M. Flentje, C. T. Supuran, Hypoxia induced CA9 inhibitory targeting by two different sulfonamide derivatives including acetazolamide in human glioblastoma. Bioorg. Med. Chem. 21, 3949–3957 (2013).

38. A. Das, N. L. Banik, S. K. Ray, Modulatory effects of acetazolomide and dexamethasone on temozolomide-mediated apoptosis in human glioblastoma T98G and U87MG cells. Cancer Invest. 26, 352–358 (2008).

39. A. Amiri, P. Uyen Le, A. Moquin, G. Machkalyan, K. Petrecca, J. W. Gillard, N. Yoganathan, D. Maysinger, Inhibition of carbonic anhydrase IX in glioblastoma multiforme. Eur. J. Pharm. Biopharm. 109, 81–92 (2016).

40. C. T. Supuran, Acetazolamide for the treatment of idiopathic intracranial hypertension. Expert Rev. Neurother. 15, 851–856 (2015).

41. N. H. Boyd, K. Walker, J. Fried, J. R. Hackney, P. C. McDonald, G. A. Benavides, R. Spina, A. Audia, S. E. Scott, C. J. Libby, A. N. Tran, M. O. Bevensee, C. Griguer, S. Nozell, G. Y. Gillespie, B. Nabors, K. P. Bhat, E. E. Bar, V. Darley-Usmar, B. Xu, E. Gordon, S. J. Cooper, S. Dedhar, A. B. Hjelmeland, Addition of carbonic anhydrase 9 inhibitor SLC-0111 to temozolomide treatment delays glioblastoma growth in vivo. JCI Insight 2, e92928 (2017).

42. J. Wu, L. Li, G. Jiang, H. Zhan, N. Wang, B-cell CLL/lymphoma 3 promotes glioma cell proliferation and inhibits apoptosis through the oncogenic STAT3 pathway. Int. J. Oncol. 49, 2471–2479 (2016).

43. B. E. Johnson, T. Mazor, C. Hong, M. Barnes, K. Aihara, C. Y. McLean, S. D. Fouse, S. Yamamoto, H. Ueda, K. Tatsuno, S. Asthana, L. E. Jalbert, S. J. Nelson, A. W. Bollen, W. C. Gustafson, E. Charron, W. A. Weiss, I. V. Smirnov, J. S. Song, A. B. Olshen, S. Cha, Y. Zhao, R. A. Moore, A. J. Mungall, S. J. Jones, M. Hirst, M. A. Marra, N. Saito, H. Aburatani, A. Mukasa, M. S. Berger, S. M. Chang, B. S. Taylor, J. F. Costello, Mutational analysis reveals the origin and therapy-driven evolution of recurrent glioma. Science 343, 189–193 (2014).

44. F. L. Muller, S. Colla, E. Aquilanti, V. E. Manzo, G. Genovese, J. Lee, D. Eisenson, R. Narurkar, P. Deng, L. Nezi, M. A. Lee, B. Hu, J. Hu, E. Sahin, D. Ong, E. Fletcher-Sananikone, D. Ho, L. Kwong, C. Brennan, Y. A. Wang, L. Chin, R. A. DePinho, Passenger deletions generate therapeutic vulnerabilities in cancer. Nature 488, 337–342 (2012).

45. D. Nijhawan, T. I. Zack, Y. Ren, M. R. Strickland, R. Lamothe, S. E. Schumacher, A. Tsherniak, H. C. Besche, J. Rosenbluh, S. Shehata, G. S. Cowley, B. A. Weir, A. L. Goldberg, J. P. Mesirov, D. E. Root, S. N. Bhatia, R. Beroukhim, W. C. Hahn, Cancer vulnerabilities unveiled by genomic loss. Cell 150, 842–854 (2012).

46. J. G. Cairncross, K. Ueki, M. C. Zlatescu, D. K. Lisle, D. M. Finkelstein, R. R. Hammond, J. S. Silver, P. C. Stark, D. R. Macdonald, Y. Ino, D. A. Ramsay, D. N. Louis, Specific genetic predictors of chemotherapeutic response and survival in patients with anaplastic oligodendrogliomas. J. Natl. Cancer Inst. 90, 1473–1479 (1998).

47. D. J. Brat, W. F. Seiferheld, A. Perry, E. H. Hammond, K. J. Murray, A. R. Schulsinger, M. P. Mehta, W. J. Curran; Radiation Therapy Oncology Group, Analysis of 1p, 19q, 9p, and 10q as prognostic markers for high-grade astrocytomas using fluorescence in situ hybridization on tissue microarrays from Radiation Therapy Oncology Group trials. Neuro Oncol. 6, 96–103 (2004).

48. J. S. Smith, I. Tachibana, U. Pohl, H. K. Lee, U. Thanarajasingam, B. P. Portier, K. Ueki, S. Ramaswamy, S. J. Billings, H. W. Mohrenweiser, D. N. Louis, R. B. Jenkins, A transcript map of the chromosome 19q-arm glioma tumor suppressor region. Genomics 64, 44–50 (2000).

49. C. Bettegowda, N. Agrawal, Y. Jiao, M. Sausen, L. D. Wood, R. H. Hruban, F. J. Rodriguez, D. P. Cahill, R. McLendon, G. Riggins, V. E. Velculescu, S. M. Oba-Shinjo, S. K. N. Marie, B. Vogelstein, D. Bigner, H. Yan, N. Papadopoulos, K. W. Kinzler, Mutations in CIC and FUBP1 contribute to human oligodendroglioma. Science 333, 1453–1455 (2011).

50. K. Shostak, X. Zhang, P. Hubert, S. I. Göktuna, Z. Jiang, I. Klevernic, J. Hildebrand, P. Roncarati, B. Hennuy, A. Ladang, J. Somja, A. Gothot, P. Close, P. Delvenne, A. Chariot, NF-B-induced KIAA1199 promotes survival through EGFR signalling. Nat. Commun. 5, 5232 (2014).

51. R. Massoumi, S. Kuphal, C. Hellerbrand, B. Haas, P. Wild, T. Spruss, A. Pfeifer, R. Fässler, A. K. Bosserhoff, Down-regulation of CYLD expression by Snail promotes tumor progression in malignant melanoma. J. Exp. Med. 206, 221–232 (2009).

52. A. M. Wakefield, J. Soukupova, A. Montagne, J. J. Ranger, R. French, W. J. Muller, R. W. E. Clarkson, Bcl3 selectively promotes metastasis of ERBB2-driven mammary tumors. Cancer Res. 15, 745–755 (2013).

53. P. Viatour, M. Bentires-Alj, A. Chariot, V. Deregowski, L. de Leval, M.-P. Merville, V. Bours, NF-B2/p100 induces Bcl-2 expression. Leukemia 17, 1349–1356 (2003).

54. D. Kashatus, P. Cogswell, A. S. Baldwin, Expression of the Bcl-3 proto-oncogene suppresses p53 activation. Genes Dev. 20, 225–235 (2006).

55. N. M. Mansour, G. M. Bernal, L. Wu, C. D. Crawley, K. E. Cahill, D. J. Voce, I. V. Balyasnikova, W. Zhang, R. Spretz, L. Nunez, G. F. Larsen, R. R. Weichselbaum, B. Yamini, Decoy receptor DcR1 is induced in a p50/Bcl3–dependent manner and attenuates the efficacy of temozolomide. Cancer Res. 75, 2039–2048 (2015).

56. J. Haapasalo, K. Nordfors, S. Järvelä, H. Bragge, I. Rantala, A.-K. Parkkila, H. Haapasalo, S. Parkkila, Carbonic anhydrase II in the endothelium of glial tumors: A potential target for therapy. Neuro Oncol. 9, 308–313 (2007).

57. B. A. Teicher, S. D. Liu, J. T. Liu, S. A. Holden, T. S. Herman, A carbonic anhydrase inhibitor as a potential modulator of cancer therapies. Anticancer Res. 13, 1549–1556 (1993).

58. R. Bayat Mokhtari, N. Baluch, M. Ka Hon Tsui, S. Kumar, T. S. Homayouni, K. Aitken, B. Das, S. Baruchel, H. Yeger, Acetazolamide potentiates the anti-tumor potential of HDACi, MS-275, in neuroblastoma. BMC Cancer 17, 156 (2017).

59. D. Neri, C. T. Supuran, Interfering with pH regulation in tumours as a therapeutic strategy. Nat. Rev. Drug Discov. 10, 767–777 (2011).

60. B. Yamini, X. Yu, M. E. Dolan, M. H. Wu, T. E. Darga, D. W. Kufe, R. R. Weichselbaum, Inhibition of nuclear factor-B activity by temozolomide involves O6-methylguanine–induced inhibition of p65 DNA binding. Cancer Res. 67, 6889–6898 (2007).

61. A. R. Tsen, P. M. Long, H. E. Driscoll, M. T. Davies, B. A. Teasdale, P. L. Penar, W. W. Pendlebury, J. L. Spees, S. E. Lawler, M. S. Viapiano, D. M. Jaworski, Triacetin-based acetate supplementation as a chemotherapeutic adjuvant therapy in glioma. Int. J. Cancer 134, 1300–1310 (2014).

62. M. S. Nandhu, B. Hu, S. E. Cole, A. Erdreich-Epstein, D. J. Rodriguez-Gil, M. S. Viapiano, Novel paracrine modulation of Notch–DLL4 signaling by fibulin-3 promotes angiogenesis in high-grade gliomas. Cancer Res. 74, 5435–5448 (2014).

63. J. Lee, S. Kotliarova, Y. Kotliarov, A. Li, Q. Su, N. M. Donin, S. Pastorino, B. W. Purow, N. Christopher, W. Zhang, J. K. Park, H. A. Fine, Tumor stem cells derived from glioblastomas cultured in bFGF and EGF more closely mirror the phenotype and genotype of primary tumors than do serum-cultured cell lines. Cancer Cell 9, 391–403 (2006).

64. X. Lan, D. J. Jörg, F. M. G. Cavalli, L. M. Richards, L. V. Nguyen, R. J. Vanner, P. Guilhamon, L. Lee, M. M. Kushida, D. Pellacani, N. I. Park, F. J. Coutinho, H. Whetstone, H. J. Selvadurai, C. Che, B. Luu, A. Carles, M. Moksa, N. Rastegar, R. Head, S. Dolma, P. Prinos,

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 15: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

Wu et al., Sci. Transl. Med. 10, eaar2238 (2018) 4 July 2018

S C I E N C E T R A N S L A T I O N A L M E D I C I N E | R E S E A R C H A R T I C L E

15 of 15

M. D. Cusimano, S. Das, M. Bernstein, C. H. Arrowsmith, A. J. Mungall, R. A. Moore, Y. Ma, M. Gallo, M. Lupien, T. J. Pugh, M. D. Taylor, M. Hirst, C. J. Eaves, B. D. Simons, P. B. Dirks, Fate mapping of human glioblastoma reveals an invariant stem cell hierarchy. Nature 549, 227–232 (2017).

65. Y. Hu, G. K. Smyth, ELDA: Extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).

66. L. A. M. Gravendeel, M. C. M. Kouwenhoven, O. Gevaert, J. J. de Rooi, A. P. Stubbs, J. E. Duijm, A. Daemen, F. E. Bleeker, L. B. C. Bralten, N. K. Kloosterhof, B. De Moor, P. H. C. Eilers, P. J. van der Spek, J. M. Kros, P. A. E. Sillevis Smitt, M. J. van den Bent, P. J. French, Intrinsic gene expression profiles of gliomas are a better predictor of survival than histology. Cancer Res. 69, 9065–9072 (2009).

67. W. A. Freije, F. E. Castro-Vargas, Z. Fang, S. Horvath, T. Cloughesy, L. M. Liau, P. S. Mischel, S. F. Nelson, Gene expression profiling of gliomas strongly predicts survival. Cancer Res. 64, 6503–6510 (2004).

68. A. Joy, A. Ramesh, I. Smirnov, M. Reiser, A. Misra, W. R. Shapiro, G. B. Mills, S. Kim, B. G. Feuerstein, AKT pathway genes define 5 prognostic subgroups in glioblastoma. PLOS ONE 9, e100827 (2014).

69. Y. Lee, A. C. Scheck, T. F. Cloughesy, A. Lai, J. Dong, H. K. Farooqi, L. M. Liau, S. Horvath, P. S. Mischel, S. F. Nelson, Gene expression analysis of glioblastomas identifies the major molecular basis for the prognostic benefit of younger age. BMC Med. Genomics 1, 52 (2008).

70. J. N. Rich, C. Hans, B. Jones, E. S. Iversen, R. E. McLendon, B. K. Ahmed Rasheed, A. Dobra, H. K. Dressman, D. D. Bigner, J. R. Nevins, M. West, Gene expression profiling and genetic markers in glioblastoma survival. Cancer Res. 65, 4051–4058 (2005).

71. L. P. Petalidis, A. Oulas, M. Backlund, M. T. Wayland, L. Liu, K. Plant, L. Happerfield, T. C. Freeman, P. Poirazi, V. P. Collins, Improved grading and survival prediction of human astrocytic brain tumors by artificial neural network analysis of gene expression microarray data. Mol. Cancer Ther. 7, 1013–1024 (2008).

72. S. Turcan, D. Rohle, A. Goenka, L. A. Walsh, F. Fang, E. Yilmaz, C. Campos, A. W. M. Fabius, C. Lu, P. S. Ward, C. B. Thompson, A. Kaufman, O. Guryanova, R. Levine, A. Heguy, A. Viale,

L. G. T. Morris, J. T. Huse, I. K. Mellinghoff, T. A. Chan, IDH1 mutation is sufficient to establish the glioma hypermethylator phenotype. Nature 483, 479–483 (2012).

Acknowledgments: We thank the University of Chicago Research Computing Center for support, the University of Chicago and Jonathon Hobbs for assistance with making and staining TMAs, L. Zhang for biostatistical assistance, and A. Uppal. Funding: This work was supported by NIH grant R01CA136937 to B.Y. and the Ludwig Center for Metastasis Research; K.E.C. was a Howard Hughes Medical Institute Research Fellow. Author contributions: L.W. performed all of the in vitro and xenograft experiments except as indicated below, analyzed all the clinical data sets including all multivariate and statistical analyses, analyzed experimental data, and edited the manuscript; G.M.B. performed two xenograft experiments with U87 cells and one with GBM43S cells; K.E.C. helped perform in vitro si-BCL3 array experiment; P.P. performed IHC analysis; C.A.F. and H.M. made FISH probes and performed FISH studies; R.R.W. edited the manuscript; B.Y. conceived and planned the research, helped perform IHC and FISH studies, analyzed the data, and wrote the manuscript. Competing interests: The authors declare that they have no competing interests. Data and materials availability: Expression data from microarray studies have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus (accession GSE80729) and are openly available. In addition, data from the publicly available data sets are freely available at the locations cited for each specific database.

Submitted 24 October 2017Resubmitted 9 April 2018Accepted 15 June 2018Published 4 July 201810.1126/scitranslmed.aar2238

Citation: L. Wu, G. M. Bernal, K. E. Cahill, P. Pytel, C. A. Fitzpatrick, H. Mashek, R. R. Weichselbaum, B. Yamini, BCL3 expression promotes resistance to alkylating chemotherapy in gliomas. Sci. Transl. Med. 10, eaar2238 (2018).

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from

Page 16: BRAIN TUMORS Copyright © 2018 expression promotes ...€¦ · BRAIN TUMORS BCL3 expression promotes resistance to alkylating chemotherapy in gliomas Longtao Wu1, Giovanna M. Bernal1,

expression promotes resistance to alkylating chemotherapy in gliomasBCL3

Weichselbaum and Bakhtiar YaminiLongtao Wu, Giovanna M. Bernal, Kirk E. Cahill, Peter Pytel, Carrie A. Fitzpatrick, Heather Mashek, Ralph R.

DOI: 10.1126/scitranslmed.aar2238, eaar2238.10Sci Transl Med

suggest that analysis of BCL3 expression might be useful in determining the best therapy for treating gliomas.inhibitor acetazolamide increased sensitivity to TMZ and survival in the mouse xenograft model. The resultsBCL3-expressing gliomas. In mice injected with high BCL3-expressing human gliomas, the carbonic anhydrase II BCL3-expressing gliomas showed a poorer response to TMZ and shorter survival than did patients with low(BCL3) expression promoted resistance to TMZ by activating carbonic anhydrase II. Patients with high

. now report that high B cell CLL/lymphoma 3et algliomas, but many patients do not respond to the treatment. Wu Chemotherapy with alkylating agents, including temozolomide (TMZ), is the most effective treatment for

BCL3 dampens drug efficacy in gliomas

ARTICLE TOOLS http://stm.sciencemag.org/content/10/448/eaar2238

MATERIALSSUPPLEMENTARY http://stm.sciencemag.org/content/suppl/2018/07/02/10.448.eaar2238.DC1

CONTENTRELATED

http://stm.sciencemag.org/content/scitransmed/10/454/eaar2718.fullhttp://stm.sciencemag.org/content/scitransmed/10/430/eaao2731.fullhttp://stm.sciencemag.org/content/scitransmed/10/441/eaao4680.fullhttp://stm.sciencemag.org/content/scitransmed/10/443/eaah6816.full

REFERENCES

http://stm.sciencemag.org/content/10/448/eaar2238#BIBLThis article cites 72 articles, 22 of which you can access for free

PERMISSIONS http://www.sciencemag.org/help/reprints-and-permissions

Terms of ServiceUse of this article is subject to the

registered trademark of AAAS. is aScience Translational MedicineScience, 1200 New York Avenue NW, Washington, DC 20005. The title

(ISSN 1946-6242) is published by the American Association for the Advancement ofScience Translational Medicine

of Science. No claim to original U.S. Government WorksCopyright © 2018 The Authors, some rights reserved; exclusive licensee American Association for the Advancement

by guest on June 22, 2020http://stm

.sciencemag.org/

Dow

nloaded from