9
Companion Diagnostics and Cancer Biomarkers Changes in BAI1 and Nestin Expression Are Prognostic Indicators for Survival and Metastases in Breast Cancer and Provide Opportunities for Dual Targeted Therapies Walter Hans Meisen 1 , Samuel Dubin 1 , Steven T. Sizemore 2 , Haritha Mathsyaraja 3 , Katie Thies 3 , Norman L. Lehman 1,4 , Peter Boyer 5,6 , Alena Cristina Jaime-Ramirez 1 , J. Bradley Elder 1 , Kimerly Powell 5,6 , Arnab Chakravarti 2 , Michael C. Ostrowski 3 , and Balveen Kaur 1,2 Abstract The 2-year survival rate of patients with breast cancer brain metastases is less than 2%. Treatment options for breast cancer brain metastases are limited, and there is an unmet need to identify novel therapies for this disease. Brain angiogenesis inhibitor 1 (BAI1) is a GPCR involved in tumor angiogenesis, invasion, phagocytosis, and synaptogenesis. For the rst time, we identify that BAI1 expression is signicantly reduced in breast cancer and higher expression is associated with better patient survival. Nestin is an intermediate lament whose expression is upregulated in several cancers. We found that higher Nestin expression signicantly correlated with breast cancer lung and brain metastases, suggesting both BAI1 and Nestin can be therapeutic targets for this disease. Here, we demonstrate the ability of an oncolytic virus, 34.5ENVE, to target and kill high Nestin-expressing cells and deliver Vstat120 (extracellular fragment of BAI1). Finally, we created two ortho- topic immune-competent murine models of breast cancer brain metastases and demonstrated 34.5ENVE extended the survival of immune-competent mice bearing intracranial breast cancer tumors. Mol Cancer Ther; 14(1); 30714. Ó2014 AACR. Introduction Breast cancer is one of the leading causes of brain metastases. The 2-year survival rate of patients with breast cancer brain metastases (BCBM) is less than 2% (1). Treatment options for patients refractory to standard surgery and radiotherapy are lim- ited. These tumors are frequently resistant to conventional che- motherapeutic drugs and antibody-based therapies, which poorly penetrate the blood brain barrier (2, 3). There is an unmet need to identify novel, targeted strategies to treat this disease. Vstat120 is a cleaved and secreted fragment of brain angiogen- esis inhibitor 1 (BAI1). The loss of BAI1 expression is observed in several cancers, including glioblastoma, colorectal cancer, gastric cancer, and renal cell carcinoma (4). The reexpression of BAI1 or Vstat120 exerts potent antiangiogenic and antitumor effects in animal models of glioblastoma and renal cell carcinoma (5, 6). Surprisingly, its expression and function has not been examined in breast cancer. Nestin is an intermediate lament whose expres- sion is upregulated in several cancers (79). Nestin is also expressed in cancer stem cells, which are known to promote cancer resistance and progression (10). Here, we determined the roles of BAI1 and Nestin gene expression in breast cancer metas- tases and patient survival. We found that lower BAI1 expression correlates with poorer patient survival, and high Nestin expression is associated with an increased probability of metastases. 34.5ENVE is an oncolytic herpes simplex virus that expresses Vstat120, and its replication is driven by a cancer stem cellspecic Nestin promoter (11). 34.5ENVE has demonstrated unparalleled antitumor efcacy in murine models of glioblastoma (11). Given the roles of Nestin and BAI1 in BCBMs, we tested the ability of 34.5ENVE to target breast cancer cells in vitro. In these studies, 34.5ENVE killed breast cancer cells of varying molecular subtypes, including those known to frequently metastasize to the brain. To test the therapeutic efcacy of a Vstat120 expressing oncolytic virus (OV) in vivo, we created three new, orthotropic models of BCBM in immune-competent FVB/NJ mice. Two of these models recapitulated the biology of human intracranial BCBM. In both of these models, we found that 34.5ENVE treatment signicantly improved the survival of mice with established BCBM. 1 Department of Neurological Surgery, James Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio. 2 Department of Radiation Oncology, James Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio. 3 Department of Molecular and Cellular Biochemistry, James Compre- hensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio. 4 Department of Pathology, James Comprehensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio. 5 Small Animal Imaging Shared Resources, James Comprehen- sive Cancer Center,The Ohio State University Medical Center, Colum- bus, Ohio. 6 Department of Biomedical Informatics, James Compre- hensive Cancer Center, The Ohio State University Medical Center, Columbus, Ohio. Note: Supplementary data for this article are available at Molecular Cancer Therapeutics Online (http://mct.aacrjournals.org/). Corresponding Author: Balveen Kaur, The Ohio State University, 385BOSUCCC, 410 West 12th Avenue, Columbus, OH 43210. Phone: 614-292-3984; Fax: 614- 688-4882; E-mail: [email protected] doi: 10.1158/1535-7163.MCT-14-0659 Ó2014 American Association for Cancer Research. Molecular Cancer Therapeutics www.aacrjournals.org 307 on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

  • Upload
    others

  • View
    9

  • Download
    0

Embed Size (px)

Citation preview

Page 1: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

Companion Diagnostics and Cancer Biomarkers

Changes in BAI1 and Nestin Expression ArePrognostic Indicators for Survival and Metastasesin Breast Cancer and Provide Opportunities forDual Targeted TherapiesWalter Hans Meisen1, Samuel Dubin1, Steven T. Sizemore2, Haritha Mathsyaraja3,Katie Thies3, Norman L. Lehman1,4, Peter Boyer5,6, Alena Cristina Jaime-Ramirez1,J. Bradley Elder1, Kimerly Powell5,6, Arnab Chakravarti2, Michael C. Ostrowski3, andBalveen Kaur1,2

Abstract

The 2-year survival rate of patients with breast cancer brainmetastases is less than 2%. Treatment options for breast cancerbrain metastases are limited, and there is an unmet need toidentify novel therapies for this disease. Brain angiogenesisinhibitor 1 (BAI1) is a GPCR involved in tumor angiogenesis,invasion, phagocytosis, and synaptogenesis. For the first time,we identify that BAI1 expression is significantly reduced inbreast cancer and higher expression is associated with betterpatient survival. Nestin is an intermediate filament whoseexpression is upregulated in several cancers. We found that

higher Nestin expression significantly correlated with breastcancer lung and brain metastases, suggesting both BAI1 andNestin can be therapeutic targets for this disease. Here, wedemonstrate the ability of an oncolytic virus, 34.5ENVE, totarget and kill high Nestin-expressing cells and deliver Vstat120(extracellular fragment of BAI1). Finally, we created two ortho-topic immune-competent murine models of breast cancer brainmetastases and demonstrated 34.5ENVE extended the survivalof immune-competent mice bearing intracranial breast cancertumors. Mol Cancer Ther; 14(1); 307–14. �2014 AACR.

IntroductionBreast cancer is one of the leading causes of brain metastases.

The 2-year survival rate of patients with breast cancer brainmetastases (BCBM) is less than 2% (1). Treatment options forpatients refractory to standard surgery and radiotherapy are lim-ited. These tumors are frequently resistant to conventional che-motherapeutic drugs and antibody-based therapies, which poorlypenetrate the blood brain barrier (2, 3). There is an unmet need toidentify novel, targeted strategies to treat this disease.

Vstat120 is a cleaved and secreted fragment of brain angiogen-esis inhibitor 1 (BAI1). The loss of BAI1 expression is observed inseveral cancers, including glioblastoma, colorectal cancer, gastriccancer, and renal cell carcinoma (4). The reexpression of BAI1 orVstat120 exerts potent antiangiogenic and antitumor effects inanimal models of glioblastoma and renal cell carcinoma (5, 6).Surprisingly, its expression and function has not been examinedin breast cancer. Nestin is an intermediate filament whose expres-sion is upregulated in several cancers (7–9). Nestin is alsoexpressed in cancer stem cells, which are known to promotecancer resistance and progression (10). Here, we determined theroles of BAI1 and Nestin gene expression in breast cancer metas-tases and patient survival. We found that lower BAI1 expressioncorrelateswithpoorer patient survival, andhighNestin expressionis associated with an increased probability of metastases.

34.5ENVE is an oncolytic herpes simplex virus that expressesVstat120, and its replication is drivenby a cancer stemcell–specificNestin promoter (11). 34.5ENVE has demonstrated unparalleledantitumor efficacy in murine models of glioblastoma (11). Giventhe roles of Nestin and BAI1 in BCBMs, we tested the abilityof 34.5ENVE to target breast cancer cells in vitro. In these studies,34.5ENVEkilled breast cancer cells of varyingmolecular subtypes,including those known to frequently metastasize to the brain.To test the therapeutic efficacy of a Vstat120 expressing oncolyticvirus (OV) in vivo, we created three new, orthotropic models ofBCBM in immune-competent FVB/NJ mice. Two of these modelsrecapitulated the biology of human intracranial BCBM. In bothof these models, we found that 34.5ENVE treatment significantlyimproved the survival of mice with established BCBM.

1Department of Neurological Surgery, James Comprehensive CancerCenter, The Ohio State University Medical Center, Columbus, Ohio.2Department of Radiation Oncology, James Comprehensive CancerCenter, The Ohio State University Medical Center, Columbus, Ohio.3Department of Molecular and Cellular Biochemistry, James Compre-hensive Cancer Center, The Ohio State University Medical Center,Columbus, Ohio. 4Department of Pathology, James ComprehensiveCancer Center, The Ohio State University Medical Center, Columbus,Ohio. 5Small Animal Imaging Shared Resources, James Comprehen-sive Cancer Center, The Ohio State University Medical Center, Colum-bus, Ohio. 6Department of Biomedical Informatics, James Compre-hensive Cancer Center, The Ohio State University Medical Center,Columbus, Ohio.

Note: Supplementary data for this article are available at Molecular CancerTherapeutics Online (http://mct.aacrjournals.org/).

CorrespondingAuthor: Balveen Kaur, The Ohio State University, 385BOSUCCC,410 West 12th Avenue, Columbus, OH 43210. Phone: 614-292-3984; Fax: 614-688-4882; E-mail: [email protected]

doi: 10.1158/1535-7163.MCT-14-0659

�2014 American Association for Cancer Research.

MolecularCancerTherapeutics

www.aacrjournals.org 307

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 2: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

Materials and MethodsCell lines and viruses

Vero, DB-7, Met-1, U251-T3, MCF7, MDA-MB-231, andMDA-MB-468 cellsweremaintained inDMEMsupplementedwith10% FBS. SKBR3 cells were maintained in McCoy's 5A Mediumsupplementedwith 10%FBS. All cellswere incubated at 37�C in anatmosphere with 5% carbon dioxide and maintained with 100units of penicillin/mL and 0.1 mg of streptomycin/mL. U251 cellswere obtained from Dr. Erwin G. Van Meir (Emory University,Atlanta, GA) and authenticated by us through the University ofArizona Genetics Core in July 2013. U251-T3 cells were created inour laboratory (May 2009) as a tumorigenic clone of U251 cells byseriallypassaging these cells three times inmice (these cells havenotbeen separately authenticated). DB-7, Met-1 (murine breast can-cer), MCF7, MDA-MB-231, SKBR3, and MDA-MB-468 (humanbreast cancer) cells were obtained in December 2012 from Dr.Michael C. Ostrowski (Ohio State University, Columbus, OH) andhave not been authenticated since receipt (12, 13). Monkey kidneyepithelial derived Vero cells were obtained inApril 2005 fromDr. EAntonio Chiocca (Ohio State University, Columbus, OH). Thesecellshavenotbeenauthenticated since receipt.All cells are routinelymonitored for changes in morphology and growth rate. All cells

were negative formycoplasma. RAMBO and 34.5ENVE viruses wereprepared and titered as previously described (11).

Cell viability assaysCells were plated in 96-well plates and infected simultaneously

with 2% FBS in DMEM containing 34.5ENVE at the indicatedmultiplicity of infection (MOI). Viability was assessed asdescribed previously using a standard 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay (14).

Virus replication assayCells (500,000) plated in 6-well plates were infected with 34.5

ENVE at anMOI of 0.005. Three days later, cells and supernatantswere harvested, and the viral titers were determined via a standardplaque-forming unit (pfu) assay.

Animal surgeryAll animal experiments were performed in accordance with the

Subcommittee on Research Animal Care of The Ohio State Uni-versity guidelines and were approved by the Institutional ReviewBoard. Female FVB/N mice (6–8-week-old; The Jackson Labora-tory) were used for in vivo tumor studies. Intracranial surgeries

-2

-1

0

Invasive ductalbreast

carcinoma

Normal

BA

I1 e

xpre

ssio

n

Mean ± 95% CIP = 0.0001

n = 389

n = 61

C

BA

0

0.2

0.4

0.6

0.8

1

5,0004,0003,0002,0001,0000

DF

S p

rob

abili

ty

Days

BAI1 high

BAI1 low

P = 0.03

n = 162

n = 162

BAI1 high

BAI1 low

0.5

1

2

MC

F10

AM

CF

12A

SU

M15

9PT

SU

M22

5CW

NS

UM

52P

EH

CC

70U

AC

C81

2M

DA

MB

231

MD

AM

B46

8LY

2H

CC

1954

MD

AM

B45

3M

DA

MB

157

MD

AM

B36

1B

T20

HC

C15

00H

CC

1007

MC

F7

ZR

751

SU

M14

9PT

BT

483

SU

M19

0PT

MD

AM

B13

460

0MP

EB

T54

9M

DA

MB

436

MD

AM

B41

5A

U56

5S

UM

185P

EH

CC

1937

HC

C11

87Z

R75

BH

CC

2185

T47

DB

T47

4H

CC

3153

SU

M44

PE

ZR

7530

CA

MA

1H

CC

1569

HC

C14

28S

UM

1315

MD

AM

B43

5H

CC

38M

DA

MB

175

HC

C11

43S

KB

R3

HC

C20

2H

BL1

00H

S57

8TH

CC

2157BA

I1 e

xpre

ssio

n

Figure 1.Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression in 61 normal mammary gland and 389 invasive ductal breastcarcinoma samples in the TCGA dataset (P ¼ 0.000137896). B, Kaplan–Meier curves representing DFS in patients of the TCGA cohort with high (n ¼ 162)or low (n ¼ 162) BAI1 expression (P ¼ 0.03). C, analysis of BAI1 expression in the Neve et al. breast cancer cell line microarray database. Expressionlevels are normalized to the nontumorigenic, epithelial cell line MCF10A.

Meisen et al.

Mol Cancer Ther; 14(1) January 2015 Molecular Cancer Therapeutics308

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 3: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

were performed as previously described with stereotactic implan-tation of 100,000 DB-7, Met-1, or Mvt1 cells (11). Tumors weretreated with Hank's Balanced Salt Solution (HBSS) or 34.5ENVEvirus at the location of tumor implantation. Animals were eutha-nized when they showed signs of morbidity.

Immunohistochemistry/immunofluorescenceMouseBCBM tumorswerefixed in zinc formalin (Anatech Ltd.)

and paraffin embedded. Tumors were sectioned at 5 mm andstained using the following antibodies: anti-MECA32 (TROMA-1), anti-F4/80 (Invitrogen; MF48000), Alexa Fluor 594 (Invitro-gen). Human tumor immunohistochemistry was performedusing antigen retrieval at pH 6.0. Tumors were stained with

anti-CD163 (Leica Microsystems Novocastra), anti-CD31 (Dako;M0823), and a horseradish peroxidase–linked secondary anti-body. Specimens were visualized with 3,30-diaminobenzidine.

Image acquisitionImmunofluorescent images were acquired using a Nikon

Eclipse E800 epifluorescence microscope equipped with a Photo-metrics Coolsnap camera and Nikon Plan Fluor objectives. Meta-Vue software (Molecular Devices) was used for image acquisition.Immunohistochemical stainingwas imagedusing aNikonEclipse50i microscope equipped with an Axiocam high resolution chan-nel camera (Zeiss) and Nikon Eclipse Ci microscope equippedwith a digital sight Fi2 camera system.

MRI analysisMice bearing intracranial tumors were treated with HBBS or

virus. Anatomic imaging was performed on the days indicatedusing a Gadolinium enhanced T1-weighted imaging sequence.For data analysis, a region-of-interest (ROI) that included thetumor was manually outlined. Tumor volumes were calculatedfrom ROIs as previously described (11).

1

2

4

8

16

MC

F10

AM

CF

12A

HC

C15

00H

CC

70Z

R75

1S

UM

159P

TH

CC

3153

MD

AM

B46

8B

T20

MD

AM

B43

6S

UM

225C

…B

T54

9Z

R75

BH

CC

1428

SU

M52

PE

HC

C19

37H

CC

1187

HC

C19

54H

BL1

00M

DA

MB

157

MD

AM

B41

5S

UM

1315

SU

M18

5PE

T47

DS

UM

190P

TH

CC

1007

SU

M44

PE

HS

578T

BT

474

SU

M14

9PT

MD

AM

B36

1LY

2C

AM

A1

600M

PE

SK

BR

3U

AC

C81

2H

CC

38A

U56

5M

DA

MB

175

BT

483

ZR

7530

HC

C20

2M

DA

MB

231

HC

C21

85H

CC

1143

MD

AM

B13

4M

CF

7M

DA

MB

453

HC

C15

69H

CC

2157

MD

AM

B43

5

NE

S e

xpre

ssio

n

0

0.2

0.4

0.6

0.8

1

100500

Pro

bab

ility

of

met

asta

sis

to

bra

in a

nd

/or

lun

g

Metastatic event follow-up time (months)

NES high

NES low

P = 0.02

n = 83

n = 83

NES high

NES low

1.25

1.5

1.75

2

2.25

ERBB2/ER/PRnegative

Other biomarkerstatus

NE

S e

xpre

ssio

n

Mean ± 95% confidence interval

P = 2.79E-37

BA

C

Figure 2.Increased Nestin expression is associated with breast cancermetastases. A, the Bos et al. cohort was stratified bymedianNES expression (83 NES-high and 83 NES-low patients) to examine the probability of brain and lungmetastases in patients with breast cancer (P¼ 0.02). B,NES expression correlateswith TNBC status in theCurtis et al. dataset. A total of 1,975 samples in the Curtis et al. breast cancer patient cohort were stratified by TNBC status. There are 250 TNBC and 1,725patients with other biomarker status in this cohort. Mean NES expression is significantly higher (P¼ 2.79E�37) in TNBCs. C, analysis of NES expression in the Neveet al. breast cancer cell line microarray database. Expression levels are normalized to the nontumorigenic, epithelial cell line MCF10A.

Table 1. Description of breast cancermolecular subtypes andNestin expressionin a panel of human breast cancer cells

Cell line Estrogen receptor Progesterone receptor HER2 Nestin

MDA-MB-231 þMDA-MB-468 þMCF7 þ þ þSKBR3 þ þ

BAI1 and Nestin in Breast Cancer Brain Metastases

www.aacrjournals.org Mol Cancer Ther; 14(1) January 2015 309

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 4: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

Statistical analysisThe Student t test was used to analyze changes in cell killing,

viral plaque-forming assays, and tumor volumemeasurements. AP value of <0.05was considered statistically significant. In survivalassays, Kaplan–Meier curves were plotted, and the log-rank testwas utilized to determine statistical significance. All statisticalanalyses were performed with the use of Graph Pad Prism soft-ware (version 5.01).

See Supplementary Materials and Methods for details.

ResultsBAI1 expression in reduced in breast cancer and is associatedwith patient survival

To determine the relevance of BAI1/Vstat120 in breast cancer,we analyzed patient-derived gene expression data from TheCancer Genome Atlas (TCGA). We observed a 52% reduction inBAI1 expression in invasive ductal breast carcinomas (n ¼ 389)compared with normal breast tissue (n ¼ 61; P < 0.0001;Fig. 1A; ref 15). Further analysis revealed that low BAI1 expres-sion was also associated with decreased disease-free survival(DFS; n ¼ 324; P < 0.03; Fig. 1B). An examination of BAI1expression in 50 breast cancer cell lines from the Neve andcolleagues dataset showed that BAI1 mRNA levels were reducedin 38% of breast cancer cell lines compared with the MCF10Abreast epithelial cell line (19 of 50 cell lines; Fig. 1C; ref. 16).These results suggest that the loss of BAI1 promotes breast

cancer tumorigenesis and the restoration of BAI1/Vstat120 mayhave therapeutic effects in breast cancer.

Nestin expression is upregulated in breast cancer and isassociated with metastases

Nestin is upregulated in several metastatic cancers, and itshigh expression correlates with reduced breast cancer patientsurvival (7–9). Nestin is also expressed in cancer stem cellsknown to promote cancer resistance and progression (10).Analysis of a cohort of 166 patients stratified by median Nestinexpression revealed a significant association between Nestinexpression and incidence of brain and lung metastases (n ¼164; P < 0.02; Fig. 2A; 17). Of the breast cancer subtypes, wefound that high Nestin expression was most strongly associatedwith triple-negative breast cancers (TNBC), which are highlyprone to brain metastases (Fig. 2B; ref. 8). Additional analysisof the Neve and colleagues microarray dataset showed thatNestin was upregulated in 100% of the breast cancer cell linesexamined (50 of 50; Fig. 2C; ref. 16). These results suggest thatNestin may be a strong therapeutic target for aggressive andmetastatic breast cancers.

Oncolytic virus is cytotoxic in multiple human breast cancersubtypes

34.5ENVE expresses Vstat120 and its replication is driven by aNestin promoter; thus, we that hypothesized 34.5ENVE might be

0

20

40

60

80

100

120

967248240

Cel

l via

bili

ty (

%)

Hours

MDA-MB-231

MDA-MB-468

MCF7

SKBR3

U251-T3

0

20

40

60

80

100

120

0.10.050.010.0050.0010

Cel

l via

bili

ty (

%)

MOI

U251-T3MCF7SKBR324

h48

hMDA-MB-231MDA-MB-468A

DB

0

20

40

60

80

100

120

MDA-MB-468

Cel

l vi

abili

ty (

%)

Untreated

RAMBO (ICP34.5-)

34.5ENVE (Nestin ICP34.5+)

***

C

Figure 3.Oncolytic HSV-derived therapeutics target and kill multiple breast cancer subtypes in vitro. A, the ability of 34.5ENVE to infect and replicate in a panel of humanbreast cancer cells was determined using 34.5ENVE-directed GFP expression. The human glioma line U251-T3 was used a positive control. Dose-dependent(B) and temporal (C) viability of human breast cancer cells treated with 34.5ENVE. Data shown are mean cell viability� SD. D, relative cytotoxicity of Nestin-driven34.5ENVE or control RAMBO virus at an MOI of 0.01 in MDA-MB-468 cells. Data shown are mean cell viability � SD (��� , P < 0.001).

Meisen et al.

Mol Cancer Ther; 14(1) January 2015 Molecular Cancer Therapeutics310

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 5: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

therapeutically relevant for BCBMs. To test this hypothesis, wetested the infection, replication, and cytotoxicity of 34.5ENVE in avariety of human breast cancer cell subtypes (Table 1). Over thecourse of 3 days, 34.5ENVE infected and replicated in humanbreast cancer cells, as determined by increasing virus encodedGFPexpression (Fig. 3A). In vitro cytotoxicity of human breast cancercells to 34.5ENVE infection was dose dependent and increasedwith time (Fig. 3B and C). Four days after infection at an MOI of0.05, we observed 83.4%, 75.7%, 80.6%, and 90.1% cell death inMDA-MB-231, SKBR3, MCF7, and MDA-MB-468 cells, respec-tively. Most breast cancer treatments are targeted to particularsubtypes, but 34.5ENVE killed breast cancer cells across multiplesubtypes. Importantly, we observed significant killing in theHer2þ and TNBC subtypes. TNBCs are notoriously resistantto conventional therapies, and Her2þ-targeted antibody treat-ments poorly penetrate the blood–brain barrier. As a result,25% to 55% of these patients with breast cancer will develop

brain metastases (18). These results highlight the therapeuticpotential of 34.5ENVE to treat BCBMs.

HSV-1 replication and cytotoxicity is enhanced by the viralneurovirulence gene ICP34.5 (11). To improve the safety andtargeting of the 34.5ENVE virus, the expression of ICP34.5 isdriven by a Nestin promoter. Nestin expression was increasedin all 50 of the breast cancer cell lines examined, suggesting thatit is a relevant therapeutic target for breast cancer (Fig. 2C).To determine the efficacy of Nestin-driven ICP34.5 on tumorcell killing, we compared the cytotoxicity of 34.5ENVE witha similar virus lacking Nestin-driven ICP34.5 expression(RAMBO; refs. 11, 19). We observed 54.14% increased killing inthe TNBC MDA-MB-468 cells in the Nestin-driven 34.5ENVEvirus, as compared with a virus without ICP34.5 (P < 0.001; Fig.3D). These results further support the use of 34.5ENVE for thetreatment of this disease. This is the first study to specifically useNestin expression to target breast cancer.

0

20

40

60

80

100

120

0.050.010.0050

Cel

l via

bili

ty (

%)

MOI

0

20

40

60

80

100

120

7248240

Cel

l via

bili

ty (

%)

Time

DB-7

Met-1

U251-T3

1

10

100

1,000

10,000

100,000

Tit

er (

PF

U/m

L)

U251-T3 DB-7 Met-1

*****

DCB

A Mvt1Met-1DB-7H

&E

Mac

rop

hag

e/m

icro

glia

Blo

od

ves

sels

Human BCBM biopsy

Figure 4.Characterization of three murine models of BCBM for preclinical evaluation of oncolytic HSV-1–derived therapeutics. A, representative panel of DB-7, Met-1,Mvt1, and human BCBM tumors. Human biopsy sample was stained for H&E, macrophages (CD163), endothelial cells (CD31;�100 magnification). Murine specimenswere stained for H&E, macrophages (F4/80), and endothelial cells (MECA-32; �20 magnification). B, 72-hour viral titers of DB-7, Met-1, and U251-T3 cells wereinfected with 34.5ENVE (0.005 MOI). Data shown are mean viral titers � SD [U251-T3 to DB7 (�� , P < 0.01); U251-T3 to Met-1 (���, P < 0.001)]. C, 48-hour cellviability of breast cancer cells treated with 34.5ENVE at the indicated MOIs. Data shown are mean cell viability� SD. D, temporal response of murine breast cancercells treated with 34.5ENVE at 0.01 MOI for 3 days. Data shown are mean cell viability � SD.

BAI1 and Nestin in Breast Cancer Brain Metastases

www.aacrjournals.org Mol Cancer Ther; 14(1) January 2015 311

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 6: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

Syngeneic model of BCBMs in an HSV-1–sensitive strainWhile there are several excellent models to study the biology of

brain metastasis development in immune-compromised mice,there are currently few immune-competent models to test thesafety and efficacy of its potential therapies (1). Cody and col-leagues (20) previously described an immune-competent modelof BCBM to test OVs, but in these murine breast cancer cells thevirus had limited antitumor efficacy in vitro and in vivo, suggestingthat it was not an optimal model to evaluate oncolytic HSV-derived therapeutics. For these studies, we characterized threenovel,murine breast cancer (DB-7,Met-1, andMvt1)models. The

DB-7 and Met-1 cells are derived from transgenic FVB/N miceexpressing polyoma virus middle T oncogene (PyVmT) under thecontrol of a mammary epithelium promoter (12). PyVmT servesas a surrogate for activated receptor tyrosine kinase signalingpathways, such as Her2, commonly activated in breast cancer(18). Mvt1 cells are derived from tumors of MMTV-c-myc/VEGFbitransgenic mice (21). In these studies, DB-7, Met-1, and Mvt1breast cancer cells were implanted intracranially into the brains ofFVB/NJ mice. DB-7 and Met-1 tumor borders were generallydemarcated from the normal brain parenchyma with localizedinvasion in the Met-1 tumors. Similar to patient specimens, thesetumors contained significant tumoral vascularization as well astumor-associated microglia/macrophages (Fig. 4A). Mvt1 tumorswere highly invasive and infiltrated into distant brain structures,including the ventricles and meninges. This phenotype was char-acteristic of rare leptomingeal metastases and did not resemblethe parenchymal brain metastases commonly observed inpatients (22). Histologic comparisons of patient BCBMs with allthree murine tumors indicated that DB-7– and Met-1–derivedtumors most closely recapitulated the human CNS metastases,

0

10

20

30

40

50

60

70

80

90

100

0 10 20 30 40

Days

HBSS

34.5ENVE

Virus

0

10

20

30

40

50

60

70

80

90

100

0 20 40 60 80

Su

rviv

al (%

)

Su

rviv

al (%

)

Days

HBSS

34.5ENVE

P = 0.0004P = 0.038

DC

Virus

7-BD1-teM

Oncolytic virus treatmentControl

treatment

1 d

ay b

efo

reT

X6

day

s af

ter

TX

10 d

ays

afte

r T

XA

0

20

40

60

80

100

120

2420130

Tu

mo

r vo

lum

e (m

m3 )

Days after tumor cell implantation

HBSS

34.5ENVE

Virus

B

*

Figure 5.Antitumor efficacy of 34.5ENVE in mice bearing established BCBM. A, representative T1-weighted MRI images of coronal sections of mice with Met-1 BCBMtumors treated with HBSS or 34.5ENVE. Scans were performed on the days indicated after treatment (TX; day 14 after tumor cell implantation). White arrowsindicate tumor location. B, mean tumor volumes (mm3) � SD of Met-1 BCBM tumors treated with HBSS or 34.5ENVE-treated mice (n ¼ 6 mice/group).C, Kaplan–Meier survival curve for A and B, mice treated on day 14 with HBSS or 2.0 � 105 pfu 34.5ENVE (n ¼ 6 mice/group, P ¼ 0.038). D, Kaplan–Meiersurvival curve of DB-7 BCBM tumors treated on day 7 with HBSS or 2.5 � 105 pfu 34.5ENVE (n ¼ 5 HBSS; n ¼ 7 34.5ENVE, P ¼ 0.0004).

Table 2. 34.5ENVE treatment reduces tumor volumes in Met-1 BCBM tumors byMRI

Days after implantation HBSS (mm3) 34.5ENVE (mm3) P

13 6.16 3.72 0.5120 25.60 8.66 0.1424 59.01 3.43 0.02

NOTE: Data shown are mean tumor volumes (mm3) of Met-1 BCBM tumorstreated with HBSS or 34.5ENVE from Fig. 5A–C (n ¼ 6 mice/group).

Mol Cancer Ther; 14(1) January 2015 Molecular Cancer Therapeutics312

Meisen et al.

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 7: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

and so these BCBMmodels were selected for further analysis (Fig.4A; ref. 23).

34.5ENVE is cytotoxic to murine breast cancer cells in vitroHuman tropic viruses often replicate poorly in murine cells, so

there are very few immune-competent models of cancer to studyOVs derived fromHSV-1. To determine whether we could evaluatethe therapeutic effectsof34.5ENVE in thismurineBCBMmodel,weexamined the ability of the virus to infect and replicate in the DB-7and Met-1 tumor–derived cell lines. For these assays, humanglioma cells were used as a positive control. We observed anincrease in virus-encoded GFP expression over 48 hours followinginfection at a lowMOI, consistentwith virus replication and spreadin these cells (Supplementary Fig. S1A). Quantification of virusreplication revealed that DB-7 and Met-1 murine cells supportedreplication at a similar rate compared with human glioma cells(Fig. 4B). 34.5ENVE killed tumor cells in a dose- and time-depen-dent manner at levels comparable to human glioma cells (Fig. 4Cand D). Three days following infection at an MOI of 0.01, weobserved 91.5%, 82.5%, and 88.2% cell death in DB-7,Met-1, andhuman glioma cells, respectively. We also verified these cellssecreted virally expressed Vstat120 and demonstrated improvedcytotoxicity of the ICP34.5-expressing 34.5ENVE virus (Supple-mentary Fig. S1B–C). The characterization of this HSV-1–sensitivemurinemodelwill aid in the future evaluationofpreclinical toxicityand efficacy of novel, HSV-1–derived therapeutics.

34.5ENVE treatment extends survival in vivoWe utilized MRI to noninvasively evaluate the antitumor

response of 34.5ENVE in mice with established Met-1 braintumors. Mice were treated intratumorally with a single dose ofHBSS or 34.5ENVE (n ¼ 6/group) 14 days after tumor cellimplantation (average initial tumor volume, 4.94 mm3).Figure 5A shows representative coronal T1-weighted MRI imagesfrom mice treated with PBS or 34.5ENVE 1 day before treatmentand on days 6 and 10 after treatment. The tumor volumes inmicetreated with HBBS grew rapidly and obtained an average tumorvolume of 59.01 mm3 within 10 days of treatment (Fig. 5Band Table 2). Significantly, 34.5ENVE-treated tumors showedsubstantial decreases in tumor volume (3.43-mm3 average tumorvolume 10 days after viral therapy; P < 0.02). Interestingly, weobserved initial pseudoprogression of tumors (by volume) in34.5ENVE-treated mice before tumor regression, possibly due totumor destruction and immune cell infiltration. Following thesemice over time, we observed that 34.5ENVE treatment signifi-cantly enhanced the survival of mice bearing Met-1 breast cancerbrain tumors. Control-treatedmice had amedian survival of only36 days, whereas mice receiving 34.5ENVE therapy survivedsignificantly longer (median survival, 52 days; P < 0.038; Fig.5C).We next tested the antitumor effects of 34.5ENVE in theDB-7BCBM model. Mice treated with 34.5ENVE showed a 100%increase in median survival compared with control mice withDB-7 tumors. HBSS- (n ¼ 5) and 34.5ENVE (n ¼ 7)-treated miceshowedmedian survival times of 17 and 34 days, respectively (P <0.0004; Fig. 5D).

DiscussionBCBMs continue to present a significant therapeutic chal-

lenge. A recent BCBM clinical trial with lapatinib and capeci-tabine noted that nearly a third of patients experienced at leastone severe adverse event due to toxicity (24). Conversely, OVtherapies, which are currently in clinical trials for a variety ofsolid tumor malignancies, including breast cancer and braintumors (NCT01656538, NCT02031965, NCT01174537, andNCT00794131), have proven to be safe and well tolerated. Inthis study, we identified BAI1/Vstat120 and Nestin as noveltherapeutic targets for BCBMs. We demonstrated that an OV,34.5ENVE, expressing antiangiogenic Vstat120 and ICP34.5under a Nestin promoter had significant cytotoxic effects inbreast cancer cells of varying molecular subtypes, includingHer2þ and TNBC. Significantly, we also described two novel,immune-competent murine models of BCBMs that closelyrecapitulate the human disease. Finally, we demonstrated thata single, intratumoral dose of 34.5ENVE virus significantlyenhanced the survival of mice with established metastaticbreast cancer brain tumors. The results of this study warrantfurther investigation of BAI1 and Nestin dual-targeted therapiesto treat established BCBMs.

Disclosure of Potential Conflicts of InterestNo potential conflicts of interest were disclosed.

Authors' ContributionsConception and design: W.H. Meisen, A.C. Jaime-Ramirez, A. Chakravarti,M.C. Ostrowski, B. KaurDevelopment of methodology: W.H. Meisen, A.C. Jaime-Ramirez, K. PowellAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): W.H. Meisen, S. Dubin, S.T. Sizemore, H. Mathsyar-aja, K. Thies, N.L. Lehman, P. Boyer, A.C. Jaime-Ramirez, J.B. Elder,A. ChakravartiAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis): W.H. Meisen, S. Dubin, S.T. Sizemore, P. Boyer,A.C. Jaime-Ramirez, J.B. Elder, K. Powell, M.C. Ostrowski, B. KaurWriting, review, and/or revision of the manuscript: W.H. Meisen, S. Dubin,S.T. Sizemore, K. Thies, N.L. Lehman, A.C. Jaime-Ramirez, J.B. Elder, A.Chakravarti, M.C. Ostrowski, B. KaurAdministrative, technical, or material support (i.e., reporting or organizingdata, constructing databases): W.H. MeisenStudy supervision: W.H. Meisen, M.C. Ostrowski, B. Kaur

Grant SupportThis work was supported in part by NIH grants R01NS064607,

R01CA150153, P30NS045758, and P01CA163205 (to B. Kaur), NIH grantP01CA097189 (to M.C. Ostrowski), NIH grant R01NS081125 (to N.L.Lehman), NIH grant P30CA016058 (to K. Powell), Pelotonia Fellowships toS. Dubin and A.C. Jaime-Ramirez, and generous support from the OSUCCCDepartments of Neurosurgery and Radiation Oncology (to B. Kaur).

The costs of publication of this articlewere defrayed inpart by the payment ofpage charges. This article must therefore be hereby marked advertisement inaccordance with 18 U.S.C. Section 1734 solely to indicate this fact.

Received August 6, 2014; revised October 13, 2014; accepted October 21,2014; published OnlineFirst November 5, 2014.

References1. Weil RJ, Palmieri DC, Bronder JL, Stark AM, Steeg PS. Breast cancer

metastasis to the central nervous system. Am J Pathol 2005;167:913–20.2. Fidler IJ, Balasubramanian K, Lin Q, Kim SW, Kim SJ. The brain microen-

vironment and cancer metastasis. Mol Cells 2010;30:93–8.

www.aacrjournals.org Mol Cancer Ther; 14(1) January 2015 313

BAI1 and Nestin in Breast Cancer Brain Metastases

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 8: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

3. Gonzalez-Angulo AM, Hortobagyi GN. Brain metastases and breast cancersubtypes. Onkologie 2010;33:143–4.

4. Cork SM, Van Meir EG. Emerging roles for the BAI1 protein family in theregulation of phagocytosis, synaptogenesis, neurovasculature, and tumordevelopment. J Mol Med (Berl) 2011;89:743–52.

5. Kaur B, Cork SM, Sandberg EM, Devi NS, Zhang Z, Klenotic PA, et al.Vasculostatin inhibits intracranial glioma growth and negatively regulatesin vivo angiogenesis through a CD36-dependent mechanism. Cancer Res2009;69:1212–20.

6. Kudo S, Konda R, Obara W, Kudo D, Tani K, Nakamura Y, et al. Inhibitionof tumor growth through suppression of angiogenesis by brain-specificangiogenesis inhibitor 1 gene transfer in murine renal cell carcinoma.Oncol Rep 2007;18:785–91.

7. KleebergerW, BovaGS, NielsenME,HerawiM, Chuang AY, Epstein JI, et al.Roles for the stem cell associated intermediate filament Nestin in prostatecancer migration and metastasis. Cancer Res 2007;67:9199–206.

8. Liu C, Chen B, Zhu J, Zhang R, Yao F, Jin F, et al. Clinical implicationsfor nestin protein expression in breast cancer. Cancer Sci 2010;101:815–9.

9. Apostolou P, Toloudi M, Chatziioannou M, Ioannou E, Papasotiriou I.Cancer stem cells stemness transcription factors expression correlates withbreast cancer disease stage. Curr Stem Cell Res Ther 2012;7:415–9.

10. Bao S, Wu Q, McLendon RE, Hao Y, Shi Q, Hjelmeland AB, et al. Gliomastem cells promote radioresistance by preferential activation of the DNAdamage response. Nature 2006;444:756–60.

11. Yoo JY, Haseley A, Bratasz A, Chiocca EA, Zhang J, Powell K, et al.Antitumor efficacy of 34.5ENVE: a transcriptionally retargeted and"Vstat120"-expressing oncolytic virus. Mol Ther 2012;20:287–97.

12. Borowsky AD, Namba R, Young LJ, Hunter KW, Hodgson JG, Tepper CG,et al. Syngeneic mouse mammary carcinoma cell lines: two closely relatedcell lines with divergent metastatic behavior. Clin Exp Metastasis2005;22:47–59.

13. Zabuawala T, Taffany DA, Sharma SM, Merchant A, Adair B, SrinivasanR, et al. An ets2-driven transcriptional program in tumor-associated

macrophages promotes tumor metastasis. Cancer Res 2010;70:1323–33.

14. Wojton J, Chu Z, Mathsyaraja H, Meisen WH, Denton N, Kwon CH, et al.Systemic delivery of SapC-DOPS has antiangiogenic and antitumor effectsagainst glioblastoma. Mol Ther 2013;21:1517–25.

15. Network TCGA. Comprehensive molecular portraits of human breasttumours. Nature 2012;490:61–70.

16. Neve RM,Chin K, Fridlyand J, Yeh J, Baehner FL, Fevr T, et al. A collection ofbreast cancer cell lines for the study of functionally distinct cancer subtypes.Cancer Cell 2006;10:515–27.

17. Bos PD, Zhang XH, Nadal C, Shu W, Gomis RR, Nguyen DX, et al. Genesthat mediate breast cancer metastasis to the brain. Nature 2009;459:1005–9.

18. Lin NU, Amiri-Kordestani L, Palmieri D, Liewehr DJ, Steeg PS. CNSmetastases in breast cancer: old challenge, new frontiers. Clin Cancer Res2013;19:6404–18.

19. Hardcastle J, Kurozumi K, Dmitrieva N, Sayers MP, Ahmad S,Waterman P,et al. Enhanced antitumor efficacy of vasculostatin (Vstat120) expressingoncolytic HSV-1. Mol Ther 2010;18:285–94.

20. Cody JJ, Scaturro P, Cantor AB, Yancey Gillespie G, Parker JN, Markert JM.Preclinical evaluation of oncolytic deltagamma(1)34.5 herpes simplexvirus expressing interleukin-12 for therapy of breast cancer brain metas-tases. Int J Breast Cancer 2012;2012:628697.

21. Pei XF, NobleMS, Davoli MA, Rosfjord E, Tilli MT, Furth PA, et al. Explant-cell culture of primary mammary tumors from MMTV-c-Myc transgenicmice. In Vitro Cell Dev Biol Anim 2004;40:14–21.

22. Scott BJ, Kesari S. Leptomeningeal metastases in breast cancer. Am J CancerRes 2013;3:117–26.

23. Pekmezci M, Perry A. Neuropathology of brainmetastases. Surg Neurol Int2013;4:S245–55.

24. Bachelot T, Romieu G, Campone M, Dieras V, Cropet C, Dalenc F, et al.Lapatinib plus capecitabine in patients with previously untreated brainmetastases from HER2-positive metastatic breast cancer (LANDSCAPE): asingle-group phase 2 study. Lancet Oncol 2013;14:64–71.

Mol Cancer Ther; 14(1) January 2015 Molecular Cancer Therapeutics314

Meisen et al.

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659

Page 9: Changes in BAI1 and Nestin Expression Are Prognostic ... · BAI1 expression Figure 1. Reduced BAI1 expression is associated with breast cancer patient survival. A, mean BAI1 expression

2015;14:307-314. Published OnlineFirst November 5, 2014.Mol Cancer Ther   Walter Hans Meisen, Samuel Dubin, Steven T. Sizemore, et al.   Opportunities for Dual Targeted Therapiesfor Survival and Metastases in Breast Cancer and Provide Changes in BAI1 and Nestin Expression Are Prognostic Indicators

  Updated version

  10.1158/1535-7163.MCT-14-0659doi:

Access the most recent version of this article at:

  Material

Supplementary

  http://mct.aacrjournals.org/content/suppl/2014/11/05/1535-7163.MCT-14-0659.DC1

Access the most recent supplemental material at:

   

   

  Cited articles

  http://mct.aacrjournals.org/content/14/1/307.full#ref-list-1

This article cites 24 articles, 4 of which you can access for free at:

  Citing articles

  http://mct.aacrjournals.org/content/14/1/307.full#related-urls

This article has been cited by 3 HighWire-hosted articles. Access the articles at:

   

  E-mail alerts related to this article or journal.Sign up to receive free email-alerts

  Subscriptions

Reprints and

  [email protected]

To order reprints of this article or to subscribe to the journal, contact the AACR Publications Department at

  Permissions

  Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

.http://mct.aacrjournals.org/content/14/1/307To request permission to re-use all or part of this article, use this link

on September 27, 2020. © 2015 American Association for Cancer Research. mct.aacrjournals.org Downloaded from

Published OnlineFirst November 5, 2014; DOI: 10.1158/1535-7163.MCT-14-0659