syrian hamster as a permissive immunocompetent animal ... · is the use of viral vectors, including...

8
Syrian Hamster as a Permissive Immunocompetent Animal Model for the Study of Oncolytic Adenovirus Vectors Maria A. Thomas, 1 Jacqueline F. Spencer, 1 Marie C. La Regina, 2 Debanjan Dhar, 1 Ann E. Tollefson, 1 Karoly Toth, 1 and William S.M. Wold 1 1 Molecular Microbiology and Immunology, Saint Louis University School of Medicine and 2 Division of Comparative Medicine, Washington University School of Medicine, St. Louis, Missouri Abstract Oncolytic adenoviruses represent an innovative approach to cancer therapy. These vectors are typically evaluated in immunodeficient mice with human xenograft tumors. How- ever, in addition to being immunodeficient, this model is limited because normal and cancerous mouse tissues are poorly permissive for human adenovirus replication. This prompted us to search for a model that more accurately reflects the use of oncolytic adenoviruses in cancer patients. To this end, we developed a novel Syrian hamster model that is both immunocompetent and replication-permissive. We found that human adenovirus replicates well in Syrian hamster cell lines and confirmed replication in the lungs. Oncolytic adenovirus injection showed efficacy in three different hamster tumor models. Furthermore, i.t. oncolytic adenovirus injection resulted in suppression of primary and metastatic lesions, i.t. replication and necrosis, vector entrance into the bloodstream, replication in the liver and lungs, and anti- adenovirus antibody induction. Our findings show that the Syrian hamster is a promising immunocompetent model that is permissive to human adenovirus replication in tumors as well as normal tissues. Therefore, the Syrian hamster model may become a valuable tool for the field of oncolytic adenovirus vectors in which vector safety and efficacy can be evaluated. (Cancer Res 2006; 66(3): 1270-6) Introduction Cancer is a leading cause of death worldwide, and there is a critical need for novel cancer therapies. One treatment approach is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory illness in young children. Oncolytic (replication competent) Ad vectors kill cancer cells as part of the natural Ad life cycle, by replicating and releasing progeny virions, lysing the cancer cell in the process. Ad replication is generally species specific, and human Ads replicate poorly in cells from most other species. For example, the natural site of Ad5 replication in humans is the lung, but attempts to detect Ad replication in the mouse lung have been unsuccessful (1, 2). Consequently, oncolytic Ad vectors are commonly evaluated in immunodeficient mice, which permit the growth of human xenograft tumors. However, this model does not accurately reflect how oncolytic Ad vectors might behave in humans because these mice lack an intact immune system and both normal and cancerous mouse tissues are poorly permissive for Ad replication (1–4). Therefore, there is a serious need for an immunocompetent animal model that is permissive for replication in malignant and normal tissues so that the toxicity, biodistribution, and antitumor efficacy of oncolytic Ad vectors can be accurately evaluated. In contrast to most other species examined, the Syrian hamster is permissive for human Ad replication in the lung (2). Following intranasal inoculation of Ad5 into Syrian hamsters, virus yields increased in the lungs, and histopathology consistent with pneumonia was observed (2). Therefore, we examined the Syrian hamster as an immunocompetent, replication-permissive animal model for the evaluation of oncolytic Ad vectors. Materials and Methods Cell lines. Syrian hamster cell lines (HaK and DDT1 MF-2) and the human cell line A549 [American Type Culture Collection (ATCC), Manassas, VA] were maintained in DMEM containing 10% fetal bovine serum (FBS; ref. 5). Although HaK cells were originally derived from normal kidneys, tumors formed in all animals. Additionally, HaK tumors metastasized to multiple sites and were histologically characterized as adenocarcinomas. The Syrian hamster pancreatic carcinoma cell line, PC1 (Dr. Parviz Pour, University of Nebraska Medical Center), was maintained in RPMI 1640 containing 10% FBS (5). Viruses and vectors. VRX-007 (6) is identical to Ad5, except the former lacks most of the E3 region (7) and overexpresses the E3 11.6K adenovirus death protein (ADP; ref. 8). VRX-007 gives yields equivalent to wild-type Ad5 (9). UV-inactivation of VRX-007 was done in a Bio-Rad GS Gene Linker UV chamber (Hercules, CA). Ad-GFP (E1 , E3 ) expresses GFP from the Rous sarcoma virus long terminal repeat. Virus stocks were purified on cesium chloride gradients and titered by plaque assays. Virus particle titers were determined by optical absorbance of viral DNA (10). Immunofluorescence. Cells were plated onto coverslips and infected 1 to 2 days later. Cells were fixed (3.7% paraformaldehyde in PBS), permeabilized (methanol containing 4V ,6-diamidino-2-phenylindole at 20jC), and immu- nostained (pool of E1A monoclonals, DBP antiserum, fiber monoclonal). Secondary antibodies were goat anti-rabbit IgG FITC-conjugate and goat anti-mouse IgG rhodamine-conjugate (Cappell/ICN, Aurora, OH). Western blots. Monolayers were infected and at each time point were washed thrice with cold PBS and frozen in radioimmunoprecipitation assay suspension buffer (11). Ten micrograms of protein were run on a 10% SDS-polyacrylamide gel, transferred to polyvinylidene fluoride membranes, and incubated with rabbit serum against Ad5 late proteins (ATCC; ref. 12). Single-step growth curves. Cells were plated onto 35-mm dishes and infected 1 to 2 days later. At 1 hour after infection, monolayers were washed thrice, and at the indicated time points, both monolayer and medium were harvested (5). Animals. Syrian (Golden) hamsters (Mesocricetus auratus ; 4-5 weeks old) were obtained from Harlan Sprague Dawley (Indianapolis, IN). Animals were allowed access to food and water ad libitum. Studies were approved by the Animal Care Committee (Saint Louis University) and done in accordance with institutional and federal regulations. Requests for reprints: William S.M. Wold, Department of Molecular Microbiology and Immunology, Saint Louis University School of Medicine, 1402 South Grand Boulevard, St. Louis, MO 63104. Phone: 314-977-8857; Fax: 314-977-8717; E-mail: [email protected]. I2006 American Association for Cancer Research. doi:10.1158/0008-5472.CAN-05-3497 Cancer Res 2006; 66: (3). February 1, 2006 1270 www.aacrjournals.org Priority Report Research. on October 1, 2020. © 2006 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 27-Jul-2020

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

Syrian Hamster as a Permissive Immunocompetent Animal Model for

the Study of Oncolytic Adenovirus Vectors

Maria A. Thomas,1Jacqueline F. Spencer,

1Marie C. La Regina,

2Debanjan Dhar,

1

Ann E. Tollefson,1Karoly Toth,

1and William S.M. Wold

1

1Molecular Microbiology and Immunology, Saint Louis University School of Medicine and 2Division of Comparative Medicine,Washington University School of Medicine, St. Louis, Missouri

Abstract

Oncolytic adenoviruses represent an innovative approach tocancer therapy. These vectors are typically evaluated inimmunodeficient mice with human xenograft tumors. How-ever, in addition to being immunodeficient, this model islimited because normal and cancerous mouse tissues arepoorly permissive for human adenovirus replication. Thisprompted us to search for a model that more accuratelyreflects the use of oncolytic adenoviruses in cancer patients.To this end, we developed a novel Syrian hamster model that isboth immunocompetent and replication-permissive. We foundthat human adenovirus replicates well in Syrian hamster celllines and confirmed replication in the lungs. Oncolyticadenovirus injection showed efficacy in three differenthamster tumor models. Furthermore, i.t. oncolytic adenovirusinjection resulted in suppression of primary and metastaticlesions, i.t. replication and necrosis, vector entrance into thebloodstream, replication in the liver and lungs, and anti-adenovirus antibody induction. Our findings show that theSyrian hamster is a promising immunocompetent model thatis permissive to human adenovirus replication in tumors aswell as normal tissues. Therefore, the Syrian hamster modelmay become a valuable tool for the field of oncolyticadenovirus vectors in which vector safety and efficacy canbe evaluated. (Cancer Res 2006; 66(3): 1270-6)

Introduction

Cancer is a leading cause of death worldwide, and there is acritical need for novel cancer therapies. One treatment approachis the use of viral vectors, including those based on adenovirusserotype 5 (Ad5), which generally causes a mild respiratory illnessin young children. Oncolytic (replication competent) Ad vectors killcancer cells as part of the natural Ad life cycle, by replicating andreleasing progeny virions, lysing the cancer cell in the process.Ad replication is generally species specific, and human Ads

replicate poorly in cells from most other species. For example, thenatural site of Ad5 replication in humans is the lung, but attemptsto detect Ad replication in the mouse lung have been unsuccessful(1, 2). Consequently, oncolytic Ad vectors are commonly evaluatedin immunodeficient mice, which permit the growth of humanxenograft tumors. However, this model does not accurately reflecthow oncolytic Ad vectors might behave in humans because these

mice lack an intact immune system and both normal andcancerous mouse tissues are poorly permissive for Ad replication(1–4). Therefore, there is a serious need for an immunocompetentanimal model that is permissive for replication in malignant andnormal tissues so that the toxicity, biodistribution, and antitumorefficacy of oncolytic Ad vectors can be accurately evaluated.In contrast to most other species examined, the Syrian hamster

is permissive for human Ad replication in the lung (2). Followingintranasal inoculation of Ad5 into Syrian hamsters, virus yieldsincreased in the lungs, and histopathology consistent withpneumonia was observed (2). Therefore, we examined the Syrianhamster as an immunocompetent, replication-permissive animalmodel for the evaluation of oncolytic Ad vectors.

Materials and Methods

Cell lines. Syrian hamster cell lines (HaK and DDT1 MF-2) and thehuman cell line A549 [American Type Culture Collection (ATCC), Manassas,

VA] were maintained in DMEM containing 10% fetal bovine serum (FBS;

ref. 5). Although HaK cells were originally derived from normal kidneys,tumors formed in all animals. Additionally, HaK tumors metastasized to

multiple sites and were histologically characterized as adenocarcinomas.

The Syrian hamster pancreatic carcinoma cell line, PC1 (Dr. Parviz Pour,

University of Nebraska Medical Center), was maintained in RPMI 1640containing 10% FBS (5).

Viruses and vectors. VRX-007 (6) is identical to Ad5, except the former

lacks most of the E3 region (7) and overexpresses the E3 11.6K adenovirus

death protein (ADP; ref. 8). VRX-007 gives yields equivalent to wild-type Ad5(9). UV-inactivation of VRX-007 was done in a Bio-Rad GS Gene Linker UV

chamber (Hercules, CA). Ad-GFP (E1�, E3�) expresses GFP from the Rous

sarcoma virus long terminal repeat. Virus stocks were purified on cesium

chloride gradients and titered by plaque assays. Virus particle titers weredetermined by optical absorbance of viral DNA (10).

Immunofluorescence. Cells were plated onto coverslips and infected 1 to2 days later. Cells were fixed (3.7% paraformaldehyde in PBS), permeabilized(methanol containing 4V,6-diamidino-2-phenylindole at �20jC), and immu-

nostained (pool of E1A monoclonals, DBP antiserum, fiber monoclonal).

Secondary antibodies were goat anti-rabbit IgG FITC-conjugate and goat

anti-mouse IgG rhodamine-conjugate (Cappell/ICN, Aurora, OH).Western blots. Monolayers were infected and at each time point

were washed thrice with cold PBS and frozen in radioimmunoprecipitation

assay suspension buffer (11). Ten micrograms of protein were run on a 10%

SDS-polyacrylamide gel, transferred to polyvinylidene fluoride membranes,and incubated with rabbit serum against Ad5 late proteins (ATCC; ref. 12).

Single-step growth curves. Cells were plated onto 35-mm dishes and

infected 1 to 2 days later. At 1 hour after infection, monolayers were washedthrice, and at the indicated time points, both monolayer and medium were

harvested (5).

Animals. Syrian (Golden) hamsters (Mesocricetus auratus ; 4-5 weeks old)

were obtained from Harlan Sprague Dawley (Indianapolis, IN). Animalswere allowed access to food and water ad libitum . Studies were approved

by the Animal Care Committee (Saint Louis University) and done in

accordance with institutional and federal regulations.

Requests for reprints: William S.M. Wold, Department of Molecular Microbiologyand Immunology, Saint Louis University School of Medicine, 1402 South GrandBoulevard, St. Louis, MO 63104. Phone: 314-977-8857; Fax: 314-977-8717; E-mail:[email protected].

I2006 American Association for Cancer Research.doi:10.1158/0008-5472.CAN-05-3497

Cancer Res 2006; 66: (3). February 1, 2006 1270 www.aacrjournals.org

Priority Report

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

S.c. tumor efficacy studies. Tumors were formed by s.c. injection ofcells (HaK, 2 � 107; DDT1 MF-2, 1 � 107; or PC1, 1 � 106) into the

hindflank(s) of hamsters (5). Established tumors were injected i.t. Tumor

volumes were monitored with digital calipers.

Endotracheal instillation. Ad5 was administered via endotrachealinstillation into anesthetized hamsters (5). Lungs were harvested, minced,

frozen and thawed thrice, homogenized, sonicated, centrifuged, and titered

by plaque assays on A549 cells.

I.t. replication and biodistribution. Animals with single HaKtumors received one i.t. injection of 2.5 � 1011 virus particles of VRX-007

(f1 � 1010 TCID50 infectious units), 2.5 � 1011 virus particles of Ad-GFP

(f1 � 109 TCID50 infectious units), or vehicle. At the indicated time points,

animals were sacrificed and organs were collected. Organs were homo-genized, frozen and thawed thrice, sonicated, centrifuged, and titered by

TCID50 assays on 293 cells (5).

Evaluation of the immune response. Sera were obtained fromhamsters before and 13 to 14 days after i.t. injection of virus.

Immunofluorescence, immunoprecipitation, and Western blots were done

on Ad5-infected cells using these sera. Virus neutralization activity was

determined by serially diluting the sera (1:2), incubating with VRX-007[1 � 103 plaque-forming units (pfu)] for 1 hour in 96-well plates, and then

seeding A549 cells in the wells. At 8 days after infection, monolayers were

fixed and stained with crystal violet. Crystal violet was extracted with 30%

acetic acid, and absorbance was quantitated at 560 nm. Neutralizingantibody titers were determined by the dilution that inhibited cytopathic

effects by >75%.

Statistical analysis. To compare two treatments, a Student’s t test wasused. Kaplan-Meier survival analysis and log-rank statistics were used to

evaluate survival curves. For all experiments, a significance level of P < 0.05

was considered significant.

Results

Expression of Ad genes in Syrian hamster cell lines. Weinitially evaluated the ability of the oncolytic Ad vector VRX-007 toinfect Syrian hamster cancer cell lines. VRX-007 (6) is identical toAd5, except the former lacks most of the E3 region (7) andoverexpresses ADP (8). VRX-007 infected HaK cells (Syrian hamsterrenal cancer cell line) and A549 cells (highly permissive humancancer cell line) with similar efficiency, even at low multiplicities ofinfection (Fig. 1A). In another experiment with HaK and twoadditional Syrian hamster cancer cell lines [PC1 (pancreaticcarcinoma) and DDT1 MF-2 (ductus deferens leiomyosarcoma)],VRX-007 infected the majority of the cells (i.e., they expressed the AdE1A and DBP proteins at 24 hours after infection), and progressioninto late infection was observed (Ad fiber expression at 48 hours;Fig. 1B). The level of late protein expression in the hamster cell lineswas comparable with that seen in A549 cells (Fig. 1C).Ad replication in vitro.When Ad replication was examined in a

single-step growth curve, the burst size at 4 days in HaK cells wasonly 7-fold less than in human A549 cells (Fig. 1D, top). VRX-007replicated well in the hamster cell lines (PC1, HaK, and DDT1MF-2), yielding more than three logs of virus growth (Fig. 1D,bottom). Therefore, human Ad is able to infect, express viralproteins, and replicate in Syrian hamster cell lines at levelscomparable with human cells.Oncolytic Ad efficacy in hamster tumor models. After

showing Ad replication in vitro , we tested the ability of oncolyticAd vectors to inhibit s.c. hamster tumor growth. Established HaKtumors were injected with VRX-007, wild-type Ad5, UV-inactivatedVRX-007, or vehicle. VRX-007 and Ad5 reduced the mean tumorgrowth rate (Fig. 2A, left) and increased animal survival (Fig. 2A ,right) compared with UV-inactivated VRX-007 and vehicle. Oneanimal treated with VRX-007 was cured and remained tumor free

>5 months after treatment. Interestingly, we observed considerableperitumoral inflammation in animals that were injected with VRX-007 or Ad5, which is reflected in the peak of tumor volumes atabout 1 week, that resolved by about 2 weeks after injection(Fig. 2A, left). Virus capsid was not responsible for this inflam-mation, as it was not observed in tumors injected with UV-inactivated VRX-007. A similar acute tumor swelling was observedin a clinical trial following administration of an oncolytic Ad vectorin cancer patients (13).Primary s.c. HaK tumors metastasized to the lungs, kidneys, and

renal lymph nodes. In addition to suppressing the growth ofprimary tumors (Fig. 2B, top left), VRX-007 suppressed metastasis(Fig. 2B, top right and bottom), as exemplified by a 75% reductionof tumor burden in the lungs (data not shown). In fact, the lungsfrom some VRX-007–treated animals contained no metastases.Histopathologic examination confirmed a significant reduction inmetastasis to the lungs (P = 0.001) and the kidneys (P = 0.02) byVRX-007 (Fig. 2B, top right).Other hamster tumor models were also examined. VRX-007

significantly reduced the growth of DDT1 MF-2 tumors (P = 0.001;Fig. 2C). Necropsy of animals in this study revealed that animalsinjected with VRX-007 had smaller tumors and fewer lungmetastases. With a third hamster cell line, PC1, VRX-007 sup-pressed tumor growth (P < 0.05) despite the fact that initial tumorvolumes were f1 mL (Fig. 2D).Examination of tumor pathology and immune response

following i.t. oncolytic Ad injection. To learn more aboutthe mechanism of tumor suppression by Ad, we examined thehistology of tumors at study termination. Histopathologic exam-ination of s.c. HaK tumors revealed areas of necrosis in manytumors injected with VRX-007 or Ad5 but not with UV-inactivatedVRX-007 or vehicle (Fig. 3A). These necrotic areas had a centralarea of primarily acellular debris (Fig. 3A , rightmost image intumor injected with VRX-007), immediately surrounded bydegenerating tumor cells and neutrophils, and more peripherallysurrounded by macrophages and fibroblasts.We also evaluated the immune response following i.t. Ad

injection. Anti-Ad antibodies were present in sera from animalsinjected with VRX-007 and Ad5 (Fig. 3B). Based on the stainingpatterns observed (F 1-h-D-arabinofuranosylcytosine), the humoralimmune response was directed against both early and late Adproteins. We also found that hamsters generated both anti-hexonand anti-fiber antibodies (Fig. 3C). When these sera were tested forneutralizing antibodies, the Ad neutralization titers ranged from1:64 to 1:256.Ad replication in Syrian hamster lungs. Because replication

in the natural site of infection is a key feature of this model, weconfirmed the historical observation of Ad5 replication in Syrianhamster lungs (2). Following endotracheal instillation of Ad5, weobserved more than four orders of magnitude of virus growth inthe lungs between days 1 and 3, and by day 9, virus clearance hadbegun (Fig. 3D).Oncolytic Ad replication and biodistribution following i.t.

injection. To assess i.t. vector replication, a single i.t. injectionof VRX-007 (or replication-defective control, Ad-GFP) wasadministered, and virus yields from tumors, blood, liver, andlungs were determined. In tumors, Ad-GFP was steadily cleared,whereas VRX-007 remained elevated for 1 week at nearly 108

infectious units per tumor (Fig. 4A). At 14 days, there wasapproximately four orders of magnitude more VRX-007 pertumor than the replication-defective virus. Taken together with

Syrian Hamster Animal Model for Oncolytic Adenoviruses

www.aacrjournals.org 1271 Cancer Res 2006; 66: (3). February 1, 2006

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

the evidence of i.t. necrosis and efficacy with replicating (but notUV-inactivated) viruses, it seems that oncolytic vector injectionleads to i.t. viral replication and subsequent necrotic cell deathof tumor cells.We also assessed the biodistribution of VRX-007 and Ad-GFP

after i.t. injection (Fig. 4B-D). Following i.t. injection, both vectorsentered the bloodstream and reached distant sites at 1.5 hoursafter injection. Both viruses were essentially cleared from thebloodstream by 1 day after injection, with the exception of oneanimal (at 4 days) injected with VRX-007. In both the liver andlungs, the replication-defective virus was essentially cleared by

1 day, whereas VRX-007 remained elevated, suggesting replication.Thus, following i.t. injection, replication occurred in the liver andlungs as well as in the tumor.

Discussion

The field of oncolytic Ad vectors is in need of a permissiveimmunocompetent animal model because the use of immunode-ficient mice bearing human xenograft tumors fails to fully assessthe safety of replicating vectors as well as the effect of the immunesystem on the vector or the tumor. This need is reflected in the

Figure 1. Human adenovirus is able to infect, express early and late adenovirus proteins, and replicate in Syrian hamster cell lines at levels comparable with infectionsof human A549 cells. A, Syrian hamster cell line HaK and the human cell line A549 were infected with VRX-007 at multiplicities of infection (MOI ) of 5, 20, and100 pfu/cell. At 24 hours after infection monolayers were fixed and immunostained for the early Ad protein, E1A; nuclei were stained with 4V,6-diamidino-2-phenylindole(DAPI) to determine the proportion of cells that were infected. Similar results were obtained with wild-type Ad5 (data not shown). B, Syrian hamster cell lines (PC1, HaK,and DDT1 MF-2) were infected with VRX-007 at 50 pfu/cell. At 24 hours after infection, cells were fixed and immunostained for the early Ad proteins E1A andDNA-binding protein (DBP ), and the nuclei were stained with 4V,6-diamidino-2-phenylindole. At 48 hours after infection, cells were fixed and immunostained forDNA-binding protein and fiber (a late Ad protein that is a component of the capsid), and the nuclei were stained with 4V,6-diamidino-2-phenylindole. Ad5 infectionswere conducted with similar results (data not shown). C, Syrian hamster cell lines (PC1, HaK, and DDT1 MF-2) as well as human A549 cells were infected with100 pfu/cell of VRX-007 or mock infected. Cells were harvested at 1, 2, and 3 days after infection, and a Western blot was done on the lysates using a rabbit polyclonalserum against adenovirus late proteins. D, top, Syrian hamster HaK cell line and the human A549 cell line were infected with VRX-007 at a multiplicity of 100 pfu/cell.Dishes were harvested at 0, 1, 2, 4, and 6 days after infection, and virus yield was determined by plaque assay of the lysates on A549 cells. The burst size at 4 daysafter infection was 1,065 pfu/cell in HaK cells and 7,553 pfu/cell in A549 cells. Bottom, three Syrian hamster cell lines were infected with VRX-007 at a multiplicity of100 pfu/cell, and dishes were harvested at 0, 2, 4, and 6 days after infection. The virus recovered was titered by plaque assay of the lysates on A549 cells.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1272 www.aacrjournals.org

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

Figure 2. Growth of Syrian hamster cell lines HaK, DDT1 MF-2, and PC1 as s.c. tumors is suppressed by i.t. injection of the oncolytic Ad vector VRX-007. A, establishedHaK tumors (mean initial volume = 270 AL; n = 12 per group) were injected with 1 � 1010 pfu of VRX-007, wild-type Ad5, UV-inactivated VRX-007, or vehicle for sixconsecutive days. Left, mean fold increase in tumor volume of each treatment group is shown through day 43, when the animals in the vehicle and UV-inactivatedVRX-007 treatment groups were sacrificed due to tumor burden. At 43 days, many vehicle-injected tumors had a discharge that caused an apparent decrease in tumorvolume. Right, a survival curve (3 mL cutoff volume) is shown for the duration of the study. The vehicle and UV-inactivated VRX-007 groups were not significantly differentfrom each other based on log-rank analysis of the survival curves (P > 0.05). The groups treated with VRX-007 and Ad5 were each significantly different from bothvehicle and UV-inactivated groups (P < 0.01), while not being significantly different from each other (P > 0.05). B, established HaK tumors (mean initial volume = 490 AL;n = 18 per group) were injected with 1 � 1010 pfu of VRX-007 or vehicle for six consecutive days. A second round of treatment (three injections of 1 � 1010 pfu/dose) wasinitiated 16 days after the start of the first round of treatment. Top left, points, mean fold increase in tumor volume; bars, SE. The suppression of tumor growth by VRX-007was statistically significant beginning on day 8, as determined by a Student’s t test (P < 0.02). Bottom, the degree of metastasis to the lungs was examined for bothgroups. At necropsy of all animals remaining at day 36, the lungs (except one lobe that was fixed for histopathologic analysis) were inflated with an India ink solution tovisualize metastases (20); normal lung tissue stained black and the metastases remained white. Top right, the degree of pulmonary and renal metastasis for animalsinjected with vehicle or VRX-007. Boxplots were generated using SPSS statistical software. Solid black line, median; box, interquartile range (containing 50% of thevalues); whiskers, range of values, excluding outliers. Outliers are defined as cases with values that lie between 1.5 and 3 box lengths from the edge of the box (o ).Extreme outliers are defined as cases with values that lie more than three box lengths from the edge of the box (* ). C, established DDT1 MF-2 tumors (mean initialvolume = 680 AL) were injected with either VRX-007 (3 � 108 pfu/injection; n = 10 tumors) or vehicle (n = 6 tumors) for six consecutive days. Points, mean fold increase intumor volume; bars, SE. The growth of tumors injected with VRX-007 or vehicle was significantly different, as determined by a Student’s t test (P = 0.001). D, establishedPC1 tumors (mean initial volume = 950 AL; n = 9 per group) were injected with VRX-007 (1 � 1010 pfu/injection) or vehicle for six consecutive days. Points, medianfold increase in tumor volume; bars, SE. The suppression of tumor growth by VRX-007 was statistically significant beginning at 13 days after injection (P < 0.05).

Syrian Hamster Animal Model for Oncolytic Adenoviruses

www.aacrjournals.org 1273 Cancer Res 2006; 66: (3). February 1, 2006

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

recent interest in developing a permissive immunocompetentanimal model (12, 14–17).We have shown that Syrian hamster cell lines supported Ad

replication and the production of infectious progeny, with yields

within 7-fold of human cells. Other Syrian hamster cancer cell lines

exist, and it seems likely that Ad will replicate in many of these as

well. Although some replication has been reported in vitro inmurine,

cotton rat, and canine cell lines, the yields are at least two orders of

magnitude less than permissive human cell lines (12, 14–16, 18). In fact,

the burst with wild-type Ad in these murine cells was <1 pfu/cell (4).The oncolytic Ad, VRX-007, exhibited considerable antitumor

efficacy in three hamster tumor models. Furthermore, VRX-007

Figure 3. I.t. injection of VRX-007 resulted in i.t. necrosis and induction of an anti-Ad immune response. A, histopathologic analysis was done at 36 days after injectionfor HaK tumors from the experiment shown in Fig. 2B . Left, a representative vehicle-injected tumor; right, a tumor injected with VRX-007. Original magnifications,�40 (left) and �400 (right ). Open arrowheads, outer skin surface; closed arrowheads, outer edge of the s.c. tumor; arrows, area of necrosis and inflammation.The necrotic response shown in the tumor injected with VRX-007 was seen in 6 of 18 tumors. These necrotic areas were within areas of viable tumor, not in thecentral necrotic area that most tumors contained. B and C, established DDT1 MF-2 tumors were injected with VRX-007, Ad5, or vehicle for two consecutive days(5 � 109 pfu/dose), and serum was obtained before vector injection (preimmune) and 13 to 14 days after injection. B, A549 cells were infected with Ad5 [multiplicityof infection = 50, F1-h-D-arabinofuranosylcytosine (ara-C) as indicated], fixed and permeabilized at 1 day after infection, and incubated with sera obtained fromhamsters in this experiment. Anti-hamster IgG-Alexa Fluor 488 was used to visualize the results by immunofluorescence microscopy. The addition of 1-h-D-arabinofuranosylcytosine inhibits Ad DNA replication; therefore, only early Ad genes are expressed. C, left, lysates of Ad5-infected (I ) or uninfected (U ) A549 cells wereelectrophoresed and transferred to a membrane. The membrane was subsequently cut into strips, and a Western blot was done with sera obtained from animalsinjected with VRX-007, Ad5, or vehicle as indicated. Right, lysates of Ad5-infected (I ) or uninfected (U ) KB cells were immunoprecipitated with either fiber or hexonmonoclonal antibody, and then a Western blot was done with serum from an animal that was injected with VRX-007. D, wild-type Ad5 (1 � 107 pfu in 100 AL totalvolume) was administered to Syrian hamsters by endotracheal instillation. Hamsters were sacrificed, and lungs were harvested on 1, 3, and 9 days after instillation(n = 2 for 1 and 3 days after instillation, n = 1 for 9 days after instillation). Lungs were homogenized and titered by plaque assay on A549 cells, and the yields arereported as the total virus yield (pfu) per animal.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1274 www.aacrjournals.org

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

replicated in hamster tumors in vivo . Notably, efficacy and i.t.necrosis were observed with VRX-007 or Ad5 but not UV-inactivatedvirus, illustrating the importance of viral replication in antitumorefficacy. The level of Ad replication in murine and cotton rat tumorswas several orders of magnitude lower (12, 15). Furthermore, thelimited antitumor efficacy seen in the murine model seems to beimmune mediated rather than due to viral replication (15, 17).Our finding that Ad replicated in two sites of anticipated

vector replication, liver and lungs, in the Syrian hamster after i.t.

injection has not been observed in other animal models (4, 12),making the Syrian hamster model most able to evaluate vectorsafety. Although limited replication was reported in the liver ofCBA mice after i.v. administration of lethal doses of Ad (3),other mouse strains (C57BL/6) do not support Ad replication inthe liver (4), and efforts to show replication in the mouse lunghave been entirely unsuccessful (1, 2). To our knowledge, theSyrian hamster (2) and cotton rat (19) are the only two speciesthat support Ad replication in the lungs. It is crucial that an

Figure 4. I.t. injection of VRX-007 into HaK tumors resulted inviral replication in the tumor, liver, and lungs. A-D, a single i.t.injection of VRX-007 or a replication-defective virus control(Ad-GFP) was administered to hamsters with s.c. HaK tumors.Ad-GFP was used as a control for the rate of viral clearance in theabsence of replication. Hamsters were sacrificed at 1.5 hours, and1, 2, 4, 7, and 14 days after injection (n = 3 per virus per timepoint). Tumors, blood, livers, and lungs were harvested,homogenized, and titered by TCID50 assay on 293 cells. A,points, mean total virus yield (infectious units) per tumor; bars,SD. B-D, levels of VRX-007 and Ad-GFP in the blood, liver, andlungs at 1.5 hours (0.06 days) and 1, 2, 4, 7, and 14 days followingi.t. injection. The data for blood are shown as total virus yield/mL ofblood, and the data for liver and lungs are shown as total virusyield per organ. The total blood volume of hamsters in this studywas approximately 6 to 8 mL. Each point represents the yield fromone animal. The sensitivity of the assay for each organ isillustrated by the dotted line. In some cases, cytopathic effect waspresent in a few wells at the lowest dilutions, but not enough wells(<50%) were positive to calculate a titer by the Reed-Muenchmethod. Presence of cytopathic effect (+); no CPE observed(�). *, blood sample of limited volume, titer <2 � 102 infectiousunits/mL.

Syrian Hamster Animal Model for Oncolytic Adenoviruses

www.aacrjournals.org 1275 Cancer Res 2006; 66: (3). February 1, 2006

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

animal model for oncolytic Ad vectors is permissive forreplication in normal tissues to examine possible toxicitiescaused by vector replication.The Syrian hamster model is advantageous over other models

examined because it is the most permissive for human Adreplication both in vitro and in vivo . This is reflected in theantitumor efficacy and i.t. replication observed in the hamstermodel. Additionally, because normal tissues are also permissive,this model can assess vector safety. In addition, the availability ofnumerous hamster cancer cell lines and the ease of animalhandling are advantages over the cotton rat model.In conclusion, we have shown that human Ad replicates in

Syrian hamster cancer cell lines and also confirmed replicationin the lungs. Furthermore, i.t. oncolytic Ad injection resulted insuppression of primary and metastatic lesions, replication andnecrosis in the tumor, vector entrance into the bloodstream,replication in the liver and lungs, and induction of an anti-Adimmune response. Many of these events are likely to occur inhumans who have been administered oncolytic Ad vectors, such as

VRX-007. This model may lead to a better understanding of theinteractions between Ad vectors and the host, which may facilitatevector development and improve the efficacy of oncolytic Adtherapy. The Syrian hamster is a promising model for theevaluation of oncolytic Ad vectors and could become a greatbenefit to the field of Ad cancer therapy.

Acknowledgments

Received 9/28/2005; revised 12/6/2005; accepted 12/12/2005.Grant support: NIH grants CA108335 and CA81829.The costs of publication of this article were defrayed in part by the payment of page

charges. This article must therefore be hereby marked advertisement in accordancewith 18 U.S.C. Section 1734 solely to indicate this fact.

We thank Peter Krajcsi for preliminary studies, Jennifer Meyer for assistance in theanimal studies, Parviz Pour (University of Nebraska Medical Center) for the PC1 cellline, and Ed Harlow (Department of Biological Chemistry and Molecular Pharmaco-logy, Harvard Medical School, Boston, MA; anti-E1A antibody), Maurice Green(Institute for Molecular Virology, Saint Louis University, St. Louis, MO; anti-DBPantibody), and Jeffrey Engler (Department of Biochemistry and Molecular Genetics,University of Alabama at Birmingham, AL; anti-fiber antibody) for providingantibodies.

References

1. Ginsberg HS, Moldawer LL, Sehgal PB, et al. A mousemodel for investigating the molecular pathogenesis ofadenovirus pneumonia. Proc Natl Acad Sci U S A1991;88:1651–5.

2. Hjorth RN, Bonde GM, Pierzchala WA, et al. A newhamster model for adenoviral vaccination. Arch Virol1988;100:279–83.

3. Duncan SJ, Gordon FC, Gregory DW, et al. Infection ofmouse liver by human adenovirus type 5. J Gen Virol1978;40:45–61.

4. Liu TC, Hallden G, Wang Y, et al. An E1B-19 kDa genedeletion mutant adenovirus demonstrates tumor necro-sis factor-enhanced cancer selectivity and enhancedoncolytic potency. Mol Ther 2004;9:786–803.

5. Thomas MA, Spencer JF, Wold WSM. The use of theSyrian hamster as an animal model for oncolyticadenovirus vectors. In: Tollefson AE, Wold WSM,editors. Adenovirus Methods and Protocols. 2nd ed.Humana Press. In press.

6. Doronin K, Toth K, Kuppuswamy M, Krajcsi P,Tollefson AE, Wold WSM. Overexpression of the ADP(E3–11.6K) protein increases cell lysis and spread ofadenovirus. Virology 2003;305:378–87.

7. Lichtenstein DL, Toth K, Doronin K, Tollefson AE,

Wold WSM. Functions and mechanisms of action ofthe adenovirus E3 proteins. Int Rev Immunol 2004;23:75–111.

8. Tollefson AE, Scaria A, Hermiston TW, Ryerse JS, WoldLJ, Wold WSM. The adenovirus death protein (E3–11.6K) is required at very late stages of infection forefficient cell lysis and release of adenovirus frominfected cells. J Virol 1996;70:2296–306.

9. Toth K, Djeha H, Ying BL, et al. An oncolyticadenovirus vector combining enhanced cell-to-cellspreading, mediated by the ADP cytolytic protein, withselective replication in cancer cells with deregulatedWnt signaling. Cancer Res 2004;64:3638–44.

10. Mittereder N, March KL, Trapnell BC. Evaluation ofthe concentration and bioactivity of adenovirus vectorsfor gene therapy. J Virol 1996;70:7498–509.

11. Tarakanova VL, Wold WS. Transforming growthfactor b1 receptor II is downregulated by E1A inadenovirus-infected cells. J Virol 2003;77:9324–36.

12. Toth K, Spencer JF, Tollefson AE, et al. Cotton rattumor model for the evaluation of oncolytic adenovi-ruses. Hum Gene Ther 2005;16:139–46.

13. Reid TR, Freeman S, Post L, McCormick F, Sze DY.Effects of Onyx-015 among metastatic colorectal cancerpatients that have failed prior treatment with 5-FU/leucovorin. Cancer Gene Ther 2005;12:673–81.

14. Bernt KM, Ni S, Tieu AT, Lieber A. Assessment of acombined, adenovirus-mediated oncolytic and immu-nostimulatory tumor therapy. Cancer Res 2005;65:4343–52.

15. Hallden G, Hill R, Wang YH, et al. Novel immuno-competent murine tumor models for the assessment ofreplication-competent oncolytic adenovirus efficacy.Mol Ther 2003;8:412–24.

16. Ternovoi VV, Le LP, Belousova N, Smith BF, Siegal GP,Curiel DT. Productive replication of human adenovirustype 5 in canine cells. J Virol 2005;79:1308–11.

17. Wang YH, Hallden G, Hill R, et al. E3 genemanipulations affect oncolytic adenovirus activity inimmunocompetent tumor models. Nat Biotechnol 2003;21:1328–35.

18. Ganly I, Mautner V, Balmain A. Productive replica-tion of human adenoviruses in mouse epidermal cells.J Virol 2000;74:2895–9.

19. Pacini DL, Dubovi EJ, Clyde WA, Jr. A new animalmodel for human respiratory tract disease due toadenovirus. J Infect Dis 1984;150:92–7.

20. Kataoka M, Schumacher G, Cristiano RJ, NeelyAtkinson E, Roth J, Mukhopadhyay T. An agent thatincreases tumor suppressor transgene product coupledwith systemic transgene delivery inhibits growth ofmetastatic lung cancer in vivo . Can Res 1998;58:4761–5.

Cancer Research

Cancer Res 2006; 66: (3). February 1, 2006 1276 www.aacrjournals.org

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: Syrian Hamster as a Permissive Immunocompetent Animal ... · is the use of viral vectors, including those based on adenovirus serotype 5 (Ad5), which generally causes a mild respiratory

2006;66:1270-1276. Cancer Res   Maria A. Thomas, Jacqueline F. Spencer, Marie C. La Regina, et al.   Model for the Study of Oncolytic Adenovirus VectorsSyrian Hamster as a Permissive Immunocompetent Animal

  Updated version

  http://cancerres.aacrjournals.org/content/66/3/1270

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/66/3/1270.full#ref-list-1

This article cites 19 articles, 9 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/66/3/1270.full#related-urls

This article has been cited by 18 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] at

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

  Permissions

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

.http://cancerres.aacrjournals.org/content/66/3/1270To request permission to re-use all or part of this article, use this link

Research. on October 1, 2020. © 2006 American Association for Cancercancerres.aacrjournals.org Downloaded from