oncolytic adenovirus coding for granulocyte macrophage colony-stimulating factor induces antitumoral...

13
Clinical Studies Oncolytic Adenovirus Coding for Granulocyte Macrophage Colony-Stimulating Factor Induces Antitumoral Immunity in Cancer Patients Vincenzo Cerullo 1,4 , Sari Pesonen 1,4 , Iulia Diaconu 1,4 , Sophie Escutenaire 1,4 , Petteri T. Arstila 2 , Matteo Ugolini 1,4 , Petri Nokisalmi 1,4 , Mari Raki 1,4 , Leena Laasonen 3 , Merja Särkioja 1,4 , Maria Rajecki 1,4 , Lotta Kangasniemi 1,4 , Kilian Guse 1,4 , Andreas Helminen 1,4,8 , Laura Ahtiainen 1,4 , Ari Ristimäki 4,5,7 , Anne Räisänen-Sokolowski 4 , Elina Haavisto 1,4 , Minna Oksanen 1,4 , Eerika Karli 1,4 , Aila Karioja-Kallio 1,4 , Sirkka-Liisa Holm 1,4 , Mauri Kouri 8 , Timo Joensuu 8 , Anna Kanerva 1,6 , and Akseli Hemminki 1,4 Abstract Granulocyte macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruiting natural killer cells and by induction of tumor-specific cytotoxic T-cells through antigen-presenting cells. On- colytic tumor cellkilling can produce a potent costimulatory danger signal and release of tumor epitopes for antigen-presenting cell sampling. Therefore, an oncolytic adenovirus coding for GMCSF was engineered and shown to induce tumor-specific immunity in an immunocompetent syngeneic hamster model. Subsequently, 20 patients with advanced solid tumors refractory to standard therapies were treated with Ad5-D24-GMCSF. Of the 16 radiologically evaluable patients, 2 had complete responses, 1 had a minor response, and 5 had disease stabilization. Responses were frequently seen in injected and noninjected tumors. Treatment was well tolerated and resulted in the induction of both tumor-specific and virus-specific immunity as measured by ELISPOT and pentamer analysis. This is the first time that oncolytic virusmediated antitumor immunity has been shown in humans. Ad5-D24-GMCSF is promising for further clinical testing. Cancer Res; 70(11); 4297309. ©2010 AACR. Introduction New approaches are needed for the treatment of metastatic solid tumors. One strategy is oncolytic viruses, which selectively replicate in and kill tumor cells (13). Replication leads to effec- tive local amplification and the virus could also enter the blood- stream for transduction of the same tumor or metastases. Oncolytic replication is an immunogenic phenomenon, and recent preclinical and clinical evidence suggests that antitumor efficacy is partially mediated by the immune re- sponse (4, 5). Adenoviruses are among the most advanced cancer gene therapy platforms and their safety and efficacy has been validated in several randomized studies (69). With regard to oncolytic adenoviruses, a few dozen trials have been performed and although there is anecdotal evidence of activity in most trials, overall single-agent efficacy has been nonimpressive. This is partially due to the attenuated nature of the few agents tested thus far. However, even such early generation viruses seem quite effective when combined with standard therapy (10). In the only randomized study performed thus far, combining oncolytic adenovirus to che- motherapy doubled the response rate (8). Adenoviruses are quite immunogenic (11), and recent pre- clinical data suggests that this might be a key aspect of their antitumor activity (4). In part because of the lack of suitable animal models, the combination of oncolytic adenoviruses and immunotherapy has not been explored in detail. Howev- er, it is known that the replication-deficient adenovirus is a useful vaccination agent due to its unparalleled capacity for gene delivery, high titer production, and immunogenicity (12). Nevertheless, because the antitumor immune response induced by adenovirus replication per se is usually not suffi- cient for clinical responses, arming oncolytic viruses with im- munostimulatory molecules is attractive. Although regularly present at tumors, the frequency and activity of CD8+ T cells is often too low to mount an effective response. This might be caused partly by the immunosup- pressive environment of tumors (13). However, preclinical data suggests that by increasing costimulatory dangersignals, immunosuppressive mechanisms could be overridden (4, 13, Authors' Affiliations: 1 Cancer Gene Therapy Group, Transplantation Laboratory, Haartman Institute and Finnish Institute of Molecular Medicine, 2 Department of Bacteriology and Immunology, Haartman Institute, 3 Helsinki Medical Imaging Center, University of Helsinki, 4 HUSLAB, Helsinki University Central Hospital, Departments of 5 Pathology and 6 Obstetrics and Gynecology, Helsinki University Central Hospital, 7 Genome Scale Biology Program, Biomedicum Helsinki, and 8 International Comprehensive Cancer Center Docrates, Helsinki, Finland Note: Supplementary data for this article are available at Cancer Research Online (http://cancerres.aacrjournals.org/). Corresponding Author: Akseli Hemminki, University of Helsinki and Helsinki University Central Hospital, P.O. Box 63, Haartmaninkatu 8, Helsinki 00014, Finland. Phone: 358-91912-5464; Fax: 358-91912-5465; E-mail: [email protected]. doi: 10.1158/0008-5472.CAN-09-3567 ©2010 American Association for Cancer Research. Cancer Research www.aacrjournals.org 4297

Upload: helsinki

Post on 08-Nov-2023

0 views

Category:

Documents


0 download

TRANSCRIPT

Clinical Studies

Cancer

Research

Oncolytic Adenovirus Coding for Granulocyte MacrophageColony-Stimulating Factor Induces Antitumoral Immunity inCancer Patients

Vincenzo Cerullo1,4, Sari Pesonen1,4, Iulia Diaconu1,4, Sophie Escutenaire1,4, Petteri T. Arstila2, Matteo Ugolini1,4,Petri Nokisalmi1,4, Mari Raki1,4, Leena Laasonen3, Merja Särkioja1,4, Maria Rajecki1,4, Lotta Kangasniemi1,4,Kilian Guse1,4, Andreas Helminen1,4,8, Laura Ahtiainen1,4, Ari Ristimäki4,5,7, Anne Räisänen-Sokolowski4,Elina Haavisto1,4, Minna Oksanen1,4, Eerika Karli1,4, Aila Karioja-Kallio1,4, Sirkka-Liisa Holm1,4, Mauri Kouri8,Timo Joensuu8, Anna Kanerva1,6, and Akseli Hemminki1,4

Abstract

Authors' ALaboratoryMedicine,Institute, 3

4HUSLAB5PathologyHospital, 7

8Internation

Note: SupResearch O

CorresponHelsinki UnHelsinki 00E-mail: aks

doi: 10.115

©2010 Am

www.aacr

Granulocyte macrophage colony-stimulating factor (GMCSF) can mediate antitumor effects by recruitingnatural killer cells and by induction of tumor-specific cytotoxic T-cells through antigen-presenting cells. On-colytic tumor cell–killing can produce a potent costimulatory danger signal and release of tumor epitopes forantigen-presenting cell sampling. Therefore, an oncolytic adenovirus coding for GMCSF was engineered andshown to induce tumor-specific immunity in an immunocompetent syngeneic hamster model. Subsequently,20 patients with advanced solid tumors refractory to standard therapies were treated with Ad5-D24-GMCSF.Of the 16 radiologically evaluable patients, 2 had complete responses, 1 had a minor response, and 5 haddisease stabilization. Responses were frequently seen in injected and noninjected tumors. Treatment was welltolerated and resulted in the induction of both tumor-specific and virus-specific immunity as measured byELISPOT and pentamer analysis. This is the first time that oncolytic virus–mediated antitumor immunity hasbeen shown in humans. Ad5-D24-GMCSF is promising for further clinical testing. Cancer Res; 70(11); 4297–309.©2010 AACR.

Introduction

New approaches are needed for the treatment of metastaticsolid tumors. One strategy is oncolytic viruses, which selectivelyreplicate in and kill tumor cells (1–3). Replication leads to effec-tive local amplification and the virus could also enter the blood-stream for transduction of the same tumor or metastases.Oncolytic replication is an immunogenic phenomenon,

and recent preclinical and clinical evidence suggests thatantitumor efficacy is partially mediated by the immune re-sponse (4, 5). Adenoviruses are among the most advancedcancer gene therapy platforms and their safety and efficacyhas been validated in several randomized studies (6–9). With

ffiliations: 1Cancer Gene Therapy Group, Transplantation, Haartman Institute and Finnish Institute of Molecular2Department of Bacteriology and Immunology, HaartmanHelsinki Medical Imaging Center, University of Helsinki,, Helsinki University Central Hospital, Departments ofand 6Obstetrics and Gynecology, Helsinki University CentralGenome Scale Biology Program, Biomedicum Helsinki, andal Comprehensive Cancer Center Docrates, Helsinki, Finland

plementary data for this article are available at Cancernline (http://cancerres.aacrjournals.org/).

ding Author: Akseli Hemminki, University of Helsinki andiversity Central Hospital, P.O. Box 63, Haartmaninkatu 8,014, Finland. Phone: 358-91912-5464; Fax: 358-91912-5465;[email protected].

8/0008-5472.CAN-09-3567

erican Association for Cancer Research.

journals.org

regard to oncolytic adenoviruses, a few dozen trials havebeen performed and although there is anecdotal evidenceof activity in most trials, overall single-agent efficacy hasbeen nonimpressive. This is partially due to the attenuatednature of the few agents tested thus far. However, even suchearly generation viruses seem quite effective when combinedwith standard therapy (10). In the only randomized studyperformed thus far, combining oncolytic adenovirus to che-motherapy doubled the response rate (8).Adenoviruses are quite immunogenic (11), and recent pre-

clinical data suggests that this might be a key aspect of theirantitumor activity (4). In part because of the lack of suitableanimal models, the combination of oncolytic adenovirusesand immunotherapy has not been explored in detail. Howev-er, it is known that the replication-deficient adenovirus is auseful vaccination agent due to its unparalleled capacity forgene delivery, high titer production, and immunogenicity(12). Nevertheless, because the antitumor immune responseinduced by adenovirus replication per se is usually not suffi-cient for clinical responses, arming oncolytic viruses with im-munostimulatory molecules is attractive.Although regularly present at tumors, the frequency and

activity of CD8+ T cells is often too low to mount an effectiveresponse. This might be caused partly by the immunosup-pressive environment of tumors (13). However, preclinicaldata suggests that by increasing costimulatory “danger” signals,immunosuppressive mechanisms could be overridden (4, 13,

4297

Cerullo et al.

4298

14). Oncolytic replication provides strong danger signals andlysis of tumor cells releases tumor-associated antigens forsampling (15). Nevertheless, this may be insufficient in theabsence of specific activation of antigen-presenting cells (13).Granulocyte macrophage colony-stimulating factor

(GMCSF) is among the most potent inducers of antitumor im-munity (16). It acts through several mechanisms, including di-rect recruitment of natural killer cells and antigen-presentingcells such as dendritic cells (17, 18). GMCSF could also specif-ically activate dendritic cells at the tumor site to increasetheir expression of costimulatory molecules to enhancecross-priming and T-cell activation rather than cross-toler-ance. Although these effects are well studied in preclinical sys-tems, and there is emerging data from cancer vaccine trials,the immunologic aspects of oncolytic viruses in cancerpatients are not well understood. For example, given the dog-ma of tumor immunosuppression, it is unknown if oncolyticviruses could induce virus- or tumor-specific T-cell responses.Here, we generated an oncolytic adenovirus armed with

GMCSF. Following preclinical testing, 20 patients with ad-vanced and treatment-refractory solid tumors were treated.This is the first time that antitumoral immunity has beenshown to result from oncolytic virus treatment of humancancer patients.

Materials and Methods

AdenovirusesAd5-D24-GMCSF was generated and amplified using stan-

dard adenovirus preparation techniques (19). Briefly, humanGMCSF (Invitrogen) was amplified with specific primers fea-turing insertion of specific restriction sites SunI/MunI.GMCSF was then subcloned into pTHSN and subsequentlyrecombined with an Ad5 rescue plasmid to generate pAd5-D24-GMCSF. This plasmid was linearized with PacI andtransfected into A549 cells for amplification and rescue.All phases of the cloning were confirmed with PCR and

multiple restriction digestions. The shuttle plasmid pTHSN-GMCSF was sequenced. The absence of wild-type E1 wasconfirmed with PCR. The E1 region, transgene, and fiberwere checked in the final virus with sequencing and PCR.To this end, viral DNA was extracted and PCR and sequenc-ing was performed to analyze the integrity of the fiber andGMCSF cDNA. All phases of the virus production were doneon A549 cells to avoid risk of wild-type recombination.GMCSF is under the E3 promoter, which results in replica-tion-associated transgene expression starting ∼8 hours afterinfection. E3 is intact except for deletion of 6.7K/gp19K.The biodistribution of adenovirus is determined by the

capsid and Ad5-based viruses have been studied extensivelypreviously (20). Toxicity studies with oncolytic Ad5-basedadenoviruses have been performed previously in mice, ham-sters, and cotton rats (20). Ad5-D24-E3 and Ad5Luc1 havebeen published previously (19).

Cell linesHamster pancreatic carcinoma–derived cell line HaP-T1

was kindly provided by Dr. Ruben Hernandez-Alcoceba

Cancer Res; 70(11) June 1, 2010

(Pamplona, Spain). The kidney hamster–derived cell lineHaK was courtesy of Prof. William S.M. Wold (St. Louis,MO). These cell lines were maintained in 10% DMEM with1% pen/strep and 1% L-glutamine. Human transformed em-bryonal kidney cell line 293 was purchased from Microbix,and human lung adenocarcinoma cell line A549 and TF1were from the American Type Culture Collection.

AnimalsAll animal protocols were reviewed and approved by the Ex-

perimental Animal Committee of the University of Helsinki andthe Provincial Government of Southern Finland. Syrian ham-sters (Mesocricetus auratus) were obtained from Taconic at 4to 5 weeks of age and quarantined for at least 1 week prior tothe study. Nudemice were purchased from Taconic. The healthstatus of the animals was frequently monitored and as soon asany sign of pain or distress was evident, they were killed.

PatientsNineteen patients with advanced metastatic tumors refrac-

tory to conventional therapies were treated with a single roundof Ad5-D24-GMCSF (Table 1). Inclusion criteria were solidtumors refractory to conventional therapies,WHOperformancescore 3 or less, and no major organ function deficiencies.Exclusion criteria were organ transplant, HIV, severe cardiovas-cular, and metabolic or pulmonary disease (e.g., symptomaticcoronary heart disease, uncontrolled blood pressure). Writteninformed consent was obtained and the study was completedaccording to Good Clinical Practice guidelines and the Declara-tion of Helsinki. This compassionate use scheme was evaluatedby theMedicolegal Department of the FinnishMinistry of SocialAffairs and Health and the Gene Technology Board.

Treatment protocolPatients received a single round of treatment with Ad5-D24-

GMCSF on day 0. Virus administration was performed by ultra-sound-guided intratumoral or intracavitary (ovarian cancer,mesothelioma) injection, and one fifth of the dose was giveni.v. The starting dose of 8 × 109 viral particles (VP) was chosenbased on safety results published by others (1, 5–9, 21). Subse-quently, the dose was escalated to 1 × 1010, 3.6 × 1010, 1 × 1011,2 × 1011, 2.5 × 1011, 3 × 1011, and 4 × 1011 (Table 2). Patients weremonitored for 24 hours in the hospital and for the following4 weeks as outpatients. Follow-up for survival was performedfor a maximum of 18 months. Physical assessment and medicalhistory were done at each visit. Side effects were recordedaccording to CTCEA 3.0. Because many cancer patients havesymptoms due to disease, preexisting symptoms were notscored if they did not become worse. However, if the symptombecame more severe, e.g., pretreatment grade 1 changed tograde 2 after treatment, it was scored as grade 2.Tumor size was assessed by contrast-enhanced computer

tomography scanning typically performed at 6 weeks. Maxi-mum tumor diameters were obtained and in some livermetastatic cases, Hounsfield units (density estimate) couldalso be counted (22). Response Evaluation Criteria in SolidTumors (RECIST) were applied to overall disease, including

Cancer Research

Engineering an Oncolytic Adenovirus Coding for GMCSF

injected and noninjected lesions. These criteria include com-plete response, partial response (>30% reduction in the sumof tumor diameters), stable disease (i.e., no response/pro-gression), and progressive disease (>20% increase; refs. 23,24). Serum tumor markers were also evaluated when elevatedat baseline. The same percentages were used except that 12%to 29% reductions are indicated as minor responses.A panel of three different proinflammatory cytokines (in-

terleukin-6, interleukin-8, and tumor necrosis factor-α) wereanalyzed by BD Cytometric Bead Array Human SolubleProtein Flex Set (Becton Dickinson) according to the instruc-tions of the manufacturer. In addition, subjective symptoms

www.aacrjournals.org

were collected by the treating physicians. For all patients,serum levels of GMCSF were also measured.

Neutralizing antibody titer293 cells were seeded at 1 × 104 cells/well on 96-well plates

and cultured overnight. Next day, cells were washed withDMEM without FCS. Serum samples were incubated at 56°Cfor 90 minutes to inactivate complement, and a 4-fold dilutionseries (1:1 to 1:16,384) was prepared in serum-free DMEM.Ad5luc1 (20) was mixed with serum dilutions and incubatedat room temperature for 30 minutes. Thereafter, cells in tripli-cates were infected with 100 VP/cell in 50 μL of mix, and

Table 1. Patients at baseline

Patient code

Age (y) Sex Diagnosis Oncologic therapies prior to Ad5/3-D24-GMCSF

Cancer Res; 70(11) June

WHO

M3

30 M Hepatocellularcancer

Surgery, 5-fluorouracil, folinic acid, cisplatin, epirubicin,gemcitabine, capecitabine, oxaliplatin, IFNα, vinorelbine,erlotinib, raltitrexed

1

C3

57 F Jejunum cancer Surgery, irinotecan, 5-fluorouracil, leucovorin, oxaliplatin,bevacizumab, capecitabine, radiation

1

R8

60 F Breast cancer 5-fluorouracil, cyclophosphamide, epirubicin, tamoxifen,docetaxel, capecitabine, paclitaxel, gemcitabine, vinorelbine,carboplatin, radiation

2

O12

28 F Ovarian cancer Surgery, docetaxel, carboplatin, paclitaxel, cisplatin, gemcitabine,doxorubicin, etoposide

1

O14

57 F Ovarian cancer Surgery, 5-fluorouracil, cyclophosphamide, epirubicin, docetaxel,paclitaxel, carboplatin, bevacizumab, erlotinib, topotecan

1

G15

71 M Gastric cancer Epirubicin, oxaliplatin, capecitabine, docetaxel 1 K18 62 M Non–small cell

lung cancer

Radiation, cisplatin, docetaxel, carboplatin, paclitaxel,

gemcitabine, erlotinib

2

T19

55 F Medullar thyroidcancer

Surgery, radiation (including 131I), dacarbazine, somatostatinanalogues, IFNα

2

M32

62 M Mesothelioma Cisplatin, erlotinib, vinorelbine 1-2 X49 60 F Cervical cancer Surgery, radiation, cisplatin, paclitaxel, carboplatin, topotecan,

bevacizumab

2

M50

60 M Mesothelioma Radiation, cisplatin, erlotinib 1 I52 58 F Melanoma Surgery, dacarbazine, vincristine, lomustine, bleomycin, IFNα,

iplimumab, radiation

2

C58

54 M Colon cancer Bevacizumab, 5-fluorouracil, leucovorin, oxaliplatin, irinotecan,capecitabine

2

R73

58 F Breast cancer Surgery, cyclophosphamide, epirubicin, 5-fluorouracil, vinorelbine,tamoxifen, anastrozole, letrozole, docetaxel

1

I78

60 F Choroidal melanoma Surgery, dacarbazine, IFNα, temozolomide, cisplatin, doxorubicin 0 O88 59 F Ovarian cancer Paclitaxel, carboplatin, doxorubicin, gemcitabine, topotecan,

vinorelbine, docetaxel, etoposide

3

U89

60 M Renal cancer Surgery, IFNα, sunitinib, sorafenib, radiation 1 S100 63 F Leiomyosarcoma Surgery, radiation, doxorubicin, ifosfamide, docetaxel,

gemcitabine, trabectedin, oxaliplatin

2

S108

42 M Synovial sarcoma Surgery, doxorubicin, ifosfamide, doxorubicin, docetaxel,paclitaxel, topotecan, bevacizumab, radiation

2

O9

61 F Ovarian cancer Paclitaxel, gemcitabine, carboplatin, doxorubicin, topotecan,Ad5/3-Cox2L-D24 2 × 1010 VP as first viral treatment

1

NOTE: Description of patient characteristics at baseline: age, sex, primary tumor, WHO performance score, and summary ofprevious therapies.

1, 2010 4299

Table 2. Summary of treatment doses, antibody titer, viral load in serum and treatment response

Neutralizing antibody titer Virus load in serum Response

Patientcode

Dose(VP)

PrimaryTumor

Week posttreatment Days posttreatment RECIST* Density/other Marker Survival

0 1 2 4 0 1 2 3–7 8–12 21–40

C3 8 × 109 Jejunum cancer 0 1,024 16,834 0 0 <500 <500 0 MR 120M3 1 × 1010 HCC 0 16,384 4,096 0 0 4,896 0 0 0 D (+5.2%) 548†

O12 3.6 × 1010 Ovarian cancer 0 16,384 16,384 0 0 0 0 0 D (+7.7%) SD 106O14 1 × 1011 Ovarian cancer 64 64 0 0 0 <500 0 0 R (−100%) CR 528†

G15 1 × 1011 Gastric cancer 1,024 16,384 16,384 0 0 565 <500 0 0 −4.6% 308†

K18 2 × 1011 NSCLC 16,384 16,384 16,384 0 <500 0 0 856 D (+15%) 59T19 2 × 1011 Thyroid cancer 0 16,384 0 765 <500 <500 0 0 D (−8.9%) MR 490†

U89 2 × 1011 Renal cancer 64 16,384 0 0 0 D (+13%) 144S100 2 × 1011 Leiomyosarcoma 0 0 16,384 0 <500 <500 D (+39%) 121S108 2 × 1011 Synovial sarcoma 0 0 256 0 <500 <500 0 D (+59%) 74M50 2.5 × 1011 Mesothelioma 256 16,384 0 0 <500 0 D (−5.7%) 403†

R8 3 × 1011 Breast cancer 64 16,384 0 <500 <500 0 R (−100%) PR 447†

M32 3 × 1011 Mesothelioma 0 256 16,384 0 0 0 0 PD‡ 125X49 3 × 1011 Cervical cancer 16 4,096 1,024 0 4,290 1,211 D (+55%) −27% 92I52 3 × 1011 Melanoma 0 256 256 0 576 D (+25%) 112I78 3 × 1011 Choroidal

melanoma0 64 0 44,876 <500 63

C58 4 × 1011 Colon cancer 256 16,384 16,384 0 1,978 4,236 D (+37%) 118R73 4 × 1011 Breast cancer 0 256 1,024 0 <500 25,787 D (−3.6%) 245†

O88 4 × 1011 Ovarian cancer 0 1,024 0 <500 Yes§ PR 126O9∥ 2 × 1011 Ovarian cancer 16,384 16,384 0 2,133¶ R (−20%) 142

NOTE: CR indicates complete disappearance of measurable tumors as defined by RECIST criteria. SD indicates stable dise , whereas PD is progressive disease (more than20% increase is sum of tumor diameters). With regard to tumor markers, CR indicates reduction of the marker to within nor l limits. MR indicates a 12% to 29% decrease inmarker, whereas PR is a more than 30% decrease. Best response indicated. SD indicates stabilization of the tumor marke no progression or response). Decrease in tumordensity has been suggested to correlate with antitumor efficacy.Abbreviations: HCC, hepatocellular carcinoma; NSCLC, non–small cell lung cancers.*Percentages indicate actual overall change in total tumor diameter.†Patients still alive at the time of follow-up cutoff.‡Percentage not measurable.§Thinning of carcinomatosis (not measurable with RECIST).∥Second treatment with oncolytic virus. Patient was treated with Ad5/3-Cox2L-D24(40) 9 weeks earlier (PD).¶Refers to Ad5-D24-GMCSF specifically.

Cerullo

etal.

Cancer

Res;

70(11)June

1,2010

CancerRese

arc

h4300

SSC

PSPPPSC

PP

PS

M

asemar (

Engineering an Oncolytic Adenovirus Coding for GMCSF

100 μL of growth medium with 10% FCS was added 1 hourlater. Twenty-four hours postinfection, cells were lysed and lu-ciferase activity was measured with Luciferase Assay System(Promega) using TopCount luminometer (Perkin-Elmer). Toevaluate the effect of neutralizing antibodies in the serum ofpatients treated with Ad5-D24-GMCSF, luciferase readingswere plotted relative to gene transfer achieved with Ad5luc1alone. The neutralizing antibody titer was determined as thelowest degree of dilution that blocked gene transfer by >80%.

Determination of virus in serum samplesTotal DNA was extracted by adding 3 μg of carrier

DNA (polydeoxyadenylic acid; Roche) to 400 μL of serumand using the QIAamp DNA mini kit. Extracted DNAwas eluted in 60 μL of nuclease-free water and DNA con-centration was measured by spectrophotometry. PCR ampli-fication was based on primers and probe targeting the E1Aregion flanking the 24 bp deletion (forward primer, 5′-TCCGGTTTCTATGCCAAACCT-3′ ; reverse primer, 5′-TCCTCCGGTGATAATGACAAGA-3′; and probe onco 5′FAM-TGATCGATCCACCCAGTGA-3′MGBNFQ). In addition,a probe complementary to a sequence included in the24 bp region targeted for deletion was used to test the sam-ples for the presence of wild-type adenovirus infection (probewt 5′VIC-TACCTGCCACGAGGCT-3′MGBNFQ).The real-time PCR conditions for each 25 μL reaction were

as follows: 2× LightCycler480 Probes Master Mix (Roche),800 nmol/L of each forward and reverse primer, 200 nmol/Lof each probe, and 250 ng of extracted DNA. PCR reactionswere carried out in a LightCycler (Roche) under the followingcycling conditions: 10 minutes at 95°C, 50 cycles of 10 secondsat 95°C, 30 seconds at 62°C, 20 seconds at 72°C, and 10 min-utes at 40°C. All samples were tested in duplicate. TaqManexogenous internal positive control reagents (Applied Biosys-tems) were used in the same PCR runs to test each sample forthe presence of PCR inhibitors. A regression standard curvewas generated using DNA extracted from serial dilutions ofAd5-D24-GMCSF (1 × 108—10 VP/mL) in normal humanserum. The limit of detection and limit of quantification forthe assay were 500 VP/mL of serum. Positive samples wereconfirmed by real-time PCR using LightCycler480 SYBR GreenI Master mix (Roche) and primers specific for adenovirusand GMCSF sequences (forward primer, 5′-AAACAC-CACCCTCCTTACCTG-3′; and reverse primer, 5′-TCATT-CATCTCAGCAGCAGTG-3′).

ELISPOT and flow cytometry analysisPeripheral blood mononuclear cells (PBMC) were isolated

by Percoll gradient according to standard protocols. Cellswere immediately frozen in CTL-CryoABC serum-free media(Cellular Technology, Ltd.) for further ELISPOT and flow cy-tometry analyses. ELISPOT was purchased as a service fromProimmune (Oxford, United Kingdom). For adenovirus ELI-SPOT, cells were stimulated with the HAdV-5 Penton peptidepool, which consists of 140 peptides, each 15 amino acidslong and overlapping by 11 amino acids. For survivin, BIRC5PONAB peptide was used. It consists of a pool of 33 peptides,each 15 amino acids long and overlapping by 11 amino acids.

www.aacrjournals.org

Flow cytometry analysis was performed on a Becton Dick-inson instrument (FACSCalibur). The following antibodieswere used: human CD3 PerCP-labeled (Becton Dickinson),human CD8 clone LT8 FITC-labeled (Proimmune), and forsurvivin, Pro5 Pentamer A*0201-LMLGEFLKL PE-labeled(Proimmune). For adenoviruses, a custom-made PE-labeledtetramer was ordered from Glycotope A*0201-GLRYRSMLL.Monoclonal antibody W6/36 (Mabtech) was used to selec-tively block MHC class I presentation.

Statistical analysisStatistical tests were performed with SPSS 15.0 and Graph-

Pad Prism version 4.00 for Mac (GraphPad Software). Mann-Whitney test was used in the animal experiments. t testswere used to assess significance in the in vitro assays.

Results

In vitro and in vivo characterization of Ad5-D24-GMCSFAd5-D24-GMCSF was generated by replacing viral gp19k

and 6.7k with GMCSF (Supplementary Fig. S1A). This strate-gy couples transgene expression with viral replication to en-sure high expression of the transgene at the tumor site (25).The virus replicates in a tumor-selective manner, thus result-ing in low systemic expression of GMCSF. The tumor speci-ficity of the virus was achieved with a 24 bp deletion, whichabrogates the retinoblastoma (Rb)-binding site of E1A. Previ-ous reports have shown that this results in selective virusreplication in cells with p16-Rb pathway defects, includingmost if not all human cancers (19).Ad5-D24-GMCSF had oncolytic potency in all tested cell

lines, and in most cases, we observed no significant differ-ences between Ad5-D24-E3 and Ad5-D24-GMCSF (Supple-mentary Fig. S2A–C). Therefore, replacement of gp19k and6.7k with GMCSF did not seem to reduce the oncolytic po-tency of the virus significantly.GMCSF secretion in vitro was seen when the virus replicat-

ed (Supplementary Fig. S3A), but not when UV-inactivatedvirus was used (data not shown). To show the functionalityof virus-derived GMCSF, we cultured the TF1 cell line (26),whose growth is strictly dependent on fully functional hu-man GMCSF, with either the commercial GMCSF or with su-pernatant from cells infected with Ad5-D24-GMCSF.Although cells cultured without GMCSF started to die atday 3, the presence of GMCSF or supernatant allowed cellsto grow (Supplementary Fig. S3B). GMCSF expression was al-so observed in tumor-bearing mice (Supplementary Fig. S3C).In accord with the in vitro results, we did not see any signif-

icant difference between Ad5-D24-E3 and Ad5-D24-GMCSFin vivo, suggesting that the expression of the transgene doesnot impair the efficacy of the virus (Supplementary Fig. S3D).Human GMCSF has no biological activity in mice and thusit did not increase the efficacy of the virus.To characterize the potency of Ad5-D24-GMCSF in an immu-

nocompetent environment, we grew syngeneic tumors in Syr-ian hamsters, which have been reported to be semi-permissivefor human adenovirus (27). Interestingly, previous data also

Cancer Res; 70(11) June 1, 2010 4301

Cerullo et al.

4302

suggests the activity of human GMCSF in hamsters (28, 29).Tumors were eradicated in animals treated with both Ad5-D24-E3 and Ad5-D24-GMCSF within 16 days (Fig. 1A).Fifteen days after tumor eradication, hamsters were chal-

lenged with the same cell line (HapT1) or a different synge-neic cell line (HaK) without further virus treatment.Interestingly, all animals in which HapT1 tumors had beeneradicated with Ad5-D24-GMCSF rejected HapT1 but nonerejected HaK tumors, suggesting that GMCSF expressionfrom the virus had enhanced tumor-specific immunity (Fig.1B and C). In animals in which HapT1 tumors had been erad-icated with Ad5-D24-E3, we observed delayed HapT1 tumorgrowth, suggesting that oncolytic adenovirus replication perse could induce a degree of immunity, but not enough to pro-tect from tumor challenge (Fig. 1B).

Cancer Res; 70(11) June 1, 2010

Safety of Ad5-D24-GMCSF in cancer patientsTreatments were generally well tolerated up to 4 × 1011

VP, and no grade 4 to 5 side effects were observed (Supple-mentary Table S1). Most patients experienced grade 1 to 2flu-like symptoms, including fever (14 of 19), chills (5 of19), fatigue (7 of 19), and injection site pain (8 of 19). PatientO88 experienced temporary worsening of intestinal function(grade 3 ileus and pain). Intestinal problems are quite com-mon in ovarian cancer patients with peritoneal metastaticdisease and this patient had similar symptoms prior to virusadministration.As the levels of certain cytokines (e.g., interleukin-6) have

been suggested to predict adenovirus-mediated toxicity (30,31), it may be of importance that only minor elevations wereseen (Supplementary Fig. S4; Supplementary Table S2).

Figure 1. Oncolytic adenoviruses eradicate pancreatic tumors in Syrian hamsters eliciting a tumor-specific immune response. A, 15 Syrian hamsters wereinoculated s.c. on both flanks with 7 × 106 HapT1 cells. Animals were treated with 1 × 109 VP on days 0, 2, and 4, growth media was used as control.Syrian hamsters have been reported to be semi-permissive for replication of human adenovirus and activity of human GMCSF. B, animals previously treatedwith Ad5-D24-E3 or Ad5-D24-GMCSF were rechallenged with the same tumor (HapT1) and tumor growth was measured over time. C, animals withHapT1 tumors previously treated with Ad5-D24-E3 or Ad5-D24-GMCSF were rechallenged with a different tumor (HaK) and tumor growth was measuredover time. For statistics, Mann-Whitney test was performed with GraphPad Prism version 4.00 for Mac (GraphPad Software, http://www.graphpad.com/;*, P < 0.05).

Cancer Research

Engineering an Oncolytic Adenovirus Coding for GMCSF

Therefore, the doses used might have been well within thetherapeutic window for this virus.

Presence of Ad5-D24-GMCSF and GMCSF in serumBecause injected virus is rapidly cleared, the extended

presence of virus genomes in serum has been suggested toindicate virus replication (32–35). All patients were negativefor Ad5-D24-GMCSF before treatment. Two days or moreafter treatment, virus was detected in the blood of 15 of18 cases (Table 2).Systemic levels of GMCSF were low, which was in accord

with the absence of significant effects on total WBC counts,suggesting effective restriction of GMCSF production to thetumor (Supplementary Fig. S4).

Neutralizing antibodiesNeutralizing antibody titers were positive in 8 of 19 cases

(42%) at baseline (Table 2). Titers increased within 2 weeksin most patients regardless of viral dose. As suggested byothers, no clear correlation was seen between neutralizingantibody titers and viral dose, antitumor activity, or toxicity(8, 32, 34, 36, 37). However, with regard to virus replicationand antibody titers, it is interesting that the antibody titerof X49 increased to only 4,096 in 1 week and decreasedthereafter, and she had high and persistent amounts ofvirus in her blood. The patient (I78) who showed the high-est serum virus load had no antibodies at baseline, and at4 weeks, had a titer of only 64. These two cases would seemto suggest that some cancer patients are indeed immuno-suppressed to some degree which compromised their anti-body production, leading to enhanced virus replication. Incontrast, patient C58 also featured high serum virus load,but had antibodies at baseline with an increase to maxi-mum in 1 week.

Efficacy of Ad5-D24-GMCSFFifteen patients were evaluable for radiologic responses

(Table 2; Fig. 2). Two patients (13%) had complete eradica-tion of all measurable disease (Fig. 2). In addition, five pa-tients (33%) had stabilization of disease progressing beforetreatment. Therefore, the clinical benefit rate was 47% ac-cording to RECIST criteria. The duration of disease stabiliza-tion was 98, 106, 490, 398, and 245 days for M3, O12, T19,M50, and R73, respectively. Disease stabilization is a surro-gate end point and thus further work is required to evaluateif this translated into actual patient benefits (38).Previous reports suggest that a decrease in tumor density

might correlate with antitumor efficacy (22). This was mea-surable in two patients, and in both cases, the density de-creased (by 4.6% and 27%; Table 2). In addition, anonmeasurable decrease of the intraperitoneal tumor wasdetected in one patient (O88). There was no detectable dif-ference in response between injected and noninjectedtumors. In fact, noninjected tumors sometimes decreasedmore clearly than injected lesions (Fig. 2A, patient M50).RECIST criteria takes into account both injected and non-injected tumors and therefore response or stabilization of dis-ease previously progressing suggests a systemic antitumor

www.aacrjournals.org

effect (23, 24). Interestingly, about one third of the treated pa-tients seemed to gain long-term survival (Fig. 2B).

Second treatment with Ad5-D24-GMCSF resulted inrapid tumor reductionEncouraged by the results in the 19 patients who received

Ad5-D24-GMCSF as their first oncolytic virus treatment, wetreated one additional patient who had received another on-colytic virus (39) 9 weeks earlier. Subsequent to Ad5-D24-GMCSF treatment, she had a 52% reduction in tumor volume(20% decrease according to RECIST) in only 17 days (Fig. 2A,patient O9). After 24 hours, there was 2,133 VP/mL of Ad5-D24-GMCSF present in her serum (Table 2).

Effect of Ad5-D24-GMCSF administration on WBCcompartmentsGMCSF levels in the serum were monitored, but increase

over baseline was observed in only three patients with a max-imum level of 70 pg/mL, suggesting a restriction of GMCSFproduction in the tumor sites. No increases in WBC wereseen (Supplementary Fig. S4). The complete phenotypic pan-el of circulating WBC was evaluated (Supplementary TableS3). Interestingly, 9 of 10 patients showed an increase inthe total number of CD8+ T cells following treatment (Sup-plementary Fig. S5). Hence, we sought to investigate whetherthese cells were specific for adenovirus or if Ad5-D24-GMCSFcould also stimulate a tumor-specific response.

Adenovirus-specific T-cell response in cancer patientstreated with Ad5-D24-GMCSFThe tumor microenvironment has been thought to be im-

munosuppressive, which might work against induction of im-munity at the tumor (15). However, because we saw anincrease in total number of CD8+ T cells, and inspired bypreclinical data (4, 13), we investigated adenovirus- andtumor-specific T-cell responses. For triggering T cell–specificimmune responses, peptides need to be presented as MHCclass I by antigen-presenting cells expressing the necessarycostimulatory molecules. Interestingly, Ad5-specific ELISPOTsuggested a strong activation of T-cell responses (Fig. 3A–D).To clarify the mechanism of the response, we blocked MHCclass I presentation with a specific antibody. As expected,this resulted in a reduction of IFN-γ secretion by ELISPOT(Fig. 3A). Therefore, an immune response developed againstadenoviruses replicating in tumors, suggesting that thetumor environment might not completely preclude theinduction of an immune response.

Tumor-specific immune responseT cells were pulsed with a peptide mix from survivin,

which is a classic tumor-associated epitope broadly ex-pressed in a variety of different tumors (ref. 40; Fig. 4A–D).In all patients analyzed, we observed a posttreatment in-crease in T cells capable of recognizing survivin. When wepretreated the PBMCs with an antibody blocking MHC classI presentation, we observed a reduction of IFN-γ secretion.To deconvolute the peptide pool used in ELISPOT, weperformed pentamer analyses in HLA-A2–positive patients

Cancer Res; 70(11) June 1, 2010 4303

Cerullo et al.

4304

Cancer Res; 70(11) June 1, 2010

Figure 2. Tumor responses and survival.A, computer tomography scan from patientstreated with Ad5-D24-GMCSF. A noninjectedtumor in mesothelioma patient M50 decreasedby 88% (overall response SD), completeresponse in patient with ovarian cancer (O14),complete response in breast cancer patient(R8), and rapid response (52% reduction) inovarian cancer patient (O9) previously treatedwith a different oncolytic adenovirus. B, overallsurvival of patients treated withAd5-D24-GMCSF.

Cancer Research

Engineering an Oncolytic Adenovirus Coding for GMCSF

Figure 3. Ad5-D24-GMCSF treatment activates adenovirus-specific CTLs. Total PBMCs were isolated from treated patients at the indicated timepoints and pulsed with an adenovirus type 5 penton-derived peptide pool. IFNγ ELISPOT was performed. Each panel represents a different patient.

W6/36 monoclonal antibody was used to selectively block MHC class I presentation.

Cancer Res; 70(11) June 1, 2010www.aacrjournals.org 4305

Cerullo et al.

4306

Figure 4. Ad5-D24-GMCSF treatment activates tumor-specific CTLs. Total PBMCs were isolated from treated patients at the indicated time points andpulsed with survivin-derived peptide pools. IFNγ ELISPOT was performed. Each panel represents a different patient. The W6/36 monoclonal antibody wasused to selectively block MHC class I presentation.

Cancer Res; 70(11) June 1, 2010 Cancer Research

Engineering an Oncolytic Adenovirus Coding for GMCSF

(Supplementary Fig. S6). PBMCs from patients before and af-ter the treatment were stained with specific antibodies forCD3, CD8, and survivin and then analyzed by flow cytometry.We observed an increase of T cells specific for survivin, in cor-relation with the results seen in ELISPOT. There were moreantiadenovirus T-cells than antisurvivin T-cells, suggestingthat the former is a stronger antigen. Thus, the majority ofthe overall induction of CD8+ cells was probably due to anti-adenovirus T-cells (Supplementary Fig. S6). Nevertheless, theclear relative increase in antisurvivin cells suggests that Ad5-D24-GMCSF was able to also induce antisurvivin reactivity.Although survivin is widely considered an ideal tumor-as-

sociated antigen (40, 41), some reports have also proposedexpression by selected normal cells (42). Therefore, whereasinduction of antisurvivin T-cells suggests tumor-specific im-munity, it does not unequivocally prove it.

Discussion

Accumulating evidence indicates that a dynamic cross-talkbetween tumors and the immune system could regulate tu-mor growth and metastasis (15, 43). Viral oncolysis can beuseful as a costimulatory danger signal for helping induceimmunity instead of tolerance towards tumor antigens (4,15). In addition, cell lysis provides an ample supply of anti-gens for sampling by antigen-presenting cells. Here, wearmed a p16-Rb pathway–selective oncolytic adenovirus withGMCSF, which could recruit natural killer cells directly to thetumor. GMCSF could also recruit dendritic cells and stimu-late them so that they can return to the lymph node for co-ordinating the cytotoxic T-cell attack on the tumor.Recombinant GMCSF has been used in humans for both

its mitogenic effect on leukocyte progenitors and for its im-munostimulating properties. It has been used as a vaccineadjuvant and as systemic agent for general antitumor im-mune stimulation (44, 45). Therefore, its safety is well estab-lished. However, systemic use for immune stimulation wascompromised by side effects related to systemic exposure,whereas efficacy may have been limited because of low localconcentrations. Thus, the production of GMCSF locally at thetumor might yield safety and efficacy benefits.Only gp16k and 6.7k were deleted from the virus genome,

therefore retaining most of E3, which is useful for effectiveoncolysis (46). Also, the 24 bp deletion used to obtain tumorselectivity might be advantageous over the most widely clin-ically used virus with an E1B55k deletion (dl1520/ONYX-015/H101), because it does not attenuate virus replication in tu-mor cells (46). It has in fact been proposed that the 24 bpconstant region 2 deletion in E1A might actually increasethe oncolytic potency of the virus (47). In contrast, replica-tion in normal tissues is minimized (1, 46).In vitro and in immunodeficient preclinical systems, Ad5-

D24-GMCSF was found to be as potent as the same viruswithout the transgene. However, in the presence of T cells,only Ad5-D24-GMCSF treatment protected animals fromsubsequent challenge with the same tumor cells. Growth ofa different cell line was not compromised by prior tumoreradication with Ad5-D24-GMCSF. These findings suggest

www.aacrjournals.org

that the expression of GMCSF by the virus (Ad5-D24-GMCSF)could induce tumor-specific immunity.Ad5-D24-GMCSF was well tolerated in patients and signs

of efficacy were seen in 63% of the patients, suggesting thatAd5-D24-GMCSF may have antitumor activity in humans.Also, one third of the patients seemed to receive a long-lasting benefit. These data are particularly intriguingconsidering that these were highly pretreated patients withadvanced and often large tumors. In this patient popula-tion, a high rate of antitumor activity and long-term sur-vival were unusual.Despite relatively conservative dosing at doses 10 to 100

fold lower than reported safe in previous studies with otheroncolytic adenoviruses (1, 5–9, 21), persistent virus sheddinginto the serum was seen in 15 of 18 patients. Systemic dis-semination of virus from the tumor might allow the trans-duction of metastases which (together with the immuneresponse) might help explain the antitumor efficacy seen ininjected and noninjected lesions. In this study, we used acombination of intravenous and intratumoral injection.However, the optimal dosing schedule has not been studiedand thus merits further investigation.One of the key findings in this article is the emphatic

cytotoxic T-cell response induced against adenovirus. AsAd5-D24-GMCSF replicates in a tumor-specific manner, thissuggests that the tumor environment is capable of mountingan immune response against tumor epitopes. However, atpresent, we cannot exclude a low degree of adenoviral geneexpression in normal tissues, which might also contribute toantiadenoviral T-cell responses.Most interestingly, Ad5-D24-GMCSF treatment resulted

in an immune response against survivin, a classic tumor-associated epitope. This suggests that the immunologic tol-erance typical of tumors could be broken with an approachcombining oncolytic virus-mediated cell killing with GMCSF-mediated recruitment and activation of dendritic cell andnatural killer cells.Efficacy gains might be obtained by combining virus with

standard treatments. For example, it has been reported thatradiation and oncolytic adenovirus could be synergistic (1).The same is true for many chemotherapeutics (10, 48).Moreover, immune suppression could yield benefits by slow-ing antibody induction or modulating the immunologictumor environment (49). Tumor load and the associated im-munosuppressive tumor environment might act against thevirus-mediated antitumor immune response. Therefore,earlier treatment might yield efficacy benefits. Furthermore,as we treated patients only once, it is possible that repeatedtreatment could increase both the antitumor immune re-sponse and the transduction of tumor masses for more ef-fective oncolysis.In summary, Ad5-D24-GMCSF seems safe for the treat-

ment of human cancer. Further clinical studies are neededto study efficacy in larger numbers of patients and to opti-mize the treatment schedule. In particular, randomized trialswould be needed to more reliably show the benefit of addingGMCSF to oncolytic adenovirus, and to assess the efficacy ofthe approach in general. Nevertheless, our data suggests that

Cancer Res; 70(11) June 1, 2010 4307

Cerullo et al.

4308

the immune tolerance of the tumor environment can be bro-ken for the induction of an antitumor T-cell response.

Disclosure of Potential Conflicts of Interest

A. Hemminki is a K. Albin Johansson Research Professor of the Foundationfor the Finnish Cancer Institute. A. Hemminki is also a founder and a share-holder in Oncos Therapeutics, Inc. The other authors disclosed no potentialconflicts of interest.

Cancer Res; 70(11) June 1, 2010

Acknowledgments

We thank Saila Eksymä-Sillman, Marina Rosliakova, Satu Nikander, KylliSkogström, Arja Vilkko, Heini Välijeesiö, Jenni Kylä-Kause, Katri Silosuo, andother Eira/Docrates personnel for help and support.

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

Received 09/26/2009; revised 03/16/2010; accepted 04/08/2010; publishedOnlineFirst 05/18/2010.

References

1. Alemany R, Balague C, Curiel DT, et al. Replicative adenoviruses for

cancer therapy. Nat Biotechnol 2000;18:723–7.2. Nemunaitis J. Live viruses in cancer treatment. Oncology (Williston

Park) 2002;16:1483–92.3. Vaha-Koskela MJ, Heikkila JE, Hinkkanen AE. Oncolytic viruses in

cancer therapy. Cancer Lett 2007;254:178–216.4. Tuve S, Liu Y, Tragoolpua K, et al. In situ adenovirus vaccination en-

gages T effector cells against cancer. Vaccine 2009;27:4225–39.5. Alemany R. A smart move against cancer for vaccinia virus. Lancet

Oncol 2008;9:507–8.6. Immonen A, Vapalahti M, Tyynela K, et al. AdvHSV-tk gene therapy

with intravenous ganciclovir improves survival in human malignantglioma: a randomised, controlled study. Mol Ther 2004;10:967–72.

7. Pearson S, Jia H, Kandachi K. China approves first gene therapy. NatBiotechnol 2004;22:3–4.

8. Yu W, Fang H. Clinical trials with oncolytic adenovirus in China. CurrCancer Drug Targets 2007;7:141–8.

9. Nemunaitis J, Clayman G, Agarwala S, et al. Biomarkers predict p53gene therapy efficacy in recurrent squamous cell carcinoma of headand neck. Mol Ther 2009;17:S139.

10. Khuri FR, Nemunaitis J, Ganly I, et al. A controlled trial of intratumoralONYX-015, a selectively-replicating adenovirus, in combination withcisplatin and 5-fluorouracil in patients with recurrent head and neckcancer. Nat Med 2000;6:879–85.

11. Cerullo V, Seiler MP, Mane V, et al. Toll-like receptor 9 triggers aninnate immune response to helper-dependent adenoviral vectors.Mol Ther 2007;15:378–85.

12. Shiver JW, Fu TM, Chen L, et al. Replication-incompetent adenoviralvaccine vector elicits effective anti-immunodeficiency-virus immuni-ty. Nature 2002;415:331–5.

13. VanOosten RL, Griffith TS. Activation of tumor-specific CD8+ T cellsafter intratumoral Ad5-TRAIL/CpG oligodeoxynucleotide combina-tion therapy. Cancer Res 2007;67:11980–90.

14. Aurisicchio L, Peruzzi D, Conforti A, et al. Treatment of mammarycarcinomas in HER-2 transgenic mice through combination of genet-ic vaccine and an agonist of Toll-like receptor 9. Clin Cancer Res2009;15:1575–84.

15. Smyth MJ, Godfrey DI, Trapani JA. A fresh look at tumor immunosur-veillance and immunotherapy. Nat Immunol 2001;2:293–9.

16. Dranoff G. GM-CSF-based cancer vaccines. Immunol Rev 2002;188:147–54.

17. Degli-Esposti MA, Smyth MJ. Close encounters of different kinds:dendritic cells and NK cells take centre stage. Nat Rev Immunol2005;5:112–24.

18. Andrews DM, Andoniou CE, Scalzo AA, et al. Cross-talk betweendendritic cells and natural killer cells in viral infection. Mol Immunol2005;42:547–55.

19. Kanerva A, Zinn KR, Chaudhuri TR, et al. Enhanced therapeutic effi-cacy for ovarian cancer with a serotype 3 receptor-targeted oncolyticadenovirus. Mol Ther 2003;8:449–58.

20. Kanerva A, Wang M, Bauerschmitz GJ, et al. Gene transfer to ovariancancer versus normal tissues with fiber-modified adenoviruses. MolTher 2002;5:695–704.

21. Nemunaitis J, Cunningham C. Emerging new therapies for chemo-therapy-resistant cancer using adenoviral vectors. Drug Resist Up-dat 2002;5:34–46.

22. Choi H, Charnsangavej C, Faria SC, et al. Correlation of computedtomography and positron emission tomography in patients with me-tastatic gastrointestinal stromal tumor treated at a single institutionwith imatinib mesylate: proposal of new computed tomography re-sponse criteria. J Clin Oncol 2007;25:1753–9.

23. Therasse P, Eisenhauer EA, Verweij J. RECIST revisited: a reviewof validation studies on tumour assessment. Eur J Cancer 2006;42:1031–9.

24. Therasse P, Le Cesne A, Van Glabbeke M, Verweij J, Judson I. RE-CIST vs. WHO: prospective comparison of response criteria in anEORTC phase II clinical trial investigating ET-743 in advanced softtissue sarcoma. Eur J Cancer 2005;41:1426–30.

25. Kanerva A, Zinn KR, Peng KW, et al. Noninvasive dual modality in vivomonitoring of the persistence and potency of a tumor targeted condi-tionally replicating adenovirus. Gene Ther 2005;12:87–94.

26. Kitamura T, Tange T, Terasawa T, et al. Establishment and charac-terization of a unique human cell line that proliferates dependently onGM-CSF, IL-3, or erythropoietin. J Cell Physiol 1989;140:323–34.

27. Thomas MA, Spencer JF, La Regina MC, et al. Syrian hamster as apermissive immunocompetent animal model for the study of oncoly-tic adenovirus vectors. Cancer Res 2006;66:1270–6.

28. Cho SA, Park JH, Seok SH, et al. Effect of granulocyte macro-phage-colony stimulating factor (GM-CSF) on 5-FU-induced ulcer-ative mucositis in hamster buccal pouches. Exp Toxicol Pathol2006;57:321–8.

29. Cohen AM, Hines DK, Korach ES, Ratzkin BJ. Activation of neutro-phil function in hamsters by recombinant human granulocyte colony-stimulating. Infect Immun 1988;56:2861–5.

30. Raper SE, Chirmule N, Lee FS, et al. Fatal systemic inflammatory re-sponse syndrome in a ornithine transcarbamylase deficient patientfollowing adenoviral gene transfer. Mol GenetMetab 2003;80:148–58.

31. Brunetti-Pierri N, Palmer DJ, Beaudet AL, Carey KD, Finegold M,Ng P. Acute toxicity after high-dose systemic injection of helper-dependent adenoviral vectors into nonhuman primates. Hum GeneTher 2004;15:35–46.

32. Nemunaitis J, Senzer N, Sarmiento S, et al. A phase I trial of intrave-nous infusion of ONYX-015 and enbrel in solid tumor patients. Can-cer Gene Ther 2007;14:885–93.

33. Nemunaitis J, Meyers T, Senzer N, et al. Phase I trial of sequentialadministration of recombinant DNA and adenovirus expressingL523S protein in early stage non-small-cell lung cancer. Mol Ther2006;13:1185–91.

34. Galanis E, Okuno S, Nascimento A, et al. Phase I-II trial of ONYX-015in combination with MAP chemotherapy in patients with advancedsarcomas. Gene Ther 2005;12:437–45.

35. Reid T, Galanis E, Abbruzzese J, et al. Hepatic arterial infusion of areplication-selective oncolytic adenovirus (dl1520): phase II viral, im-munologic, and clinical endpoints. Cancer Res 2002;62:6070–9.

36. Nemunaitis J, Vorhies JS, Pappen B, Senzer N. 10-year follow-up ofgene-modified adenoviral-based therapy in 146 non-small-cell lungcancer patients. Cancer Gene Ther 2007;14:762–3.

37. Nemunaitis J, Khuri F, Ganly I, et al. Phase II trial of intratumoral ad-ministration of ONYX-015, a replication-selective adenovirus, inpatients with refractory head and neck cancer. J Clin Oncol 2001;19:289–98.

38. Ohorodnyk P, Eisenhauer EA, Booth CM. Clinical benefit in oncology

Cancer Research

Engineering an Oncolytic Adenovirus Coding for GMCSF

trials: is this a patient-centred or tumour-centred end-point? Eur JCancer 2009;45:2249–52.

39. Pesonen S, Nokisalmi P, Escutenaire S, et al. Prolonged systemic cir-culation of chimeric oncolytic adenovirus Ad5/3-Cox2L-D24 in pa-tients with metastatic and refractory solid tumors. Gene Ther. Epub2010 Mar 18.

40. Altieri DC. Survivin, cancer networks and pathway-directed drug dis-covery. Nat Rev Cancer 2008;8:61–70.

41. Ryan BM, O'Donovan N, Duffy MJ. Survivin: a new target for anti-cancer therapy. Cancer Treat Rev 2009;35:553–62.

42. Fukuda S, Pelus LM. Survivin, a cancer target with an emerging rolein normal adult tissues. Mol Cancer Ther 2006;5:1087–98.

43. Krieg AM. Therapeutic potential of Toll-like receptor 9 activation. NatRev Drug Discov 2006;5:471–84.

44. Perales MA, Yuan J, Powel S, et al. Phase I/II study of GM-CSF DNAas an adjuvant for a multipeptide cancer vaccine in patients with ad-vanced melanoma. Mol Ther 2008;16:2022–9.

www.aacrjournals.org

45. Dessureault S, Noyes D, Lee D, et al. A phase-I trial using a universalGM-CSF-producing and CD40L-expressing bystander cell line (GMCD40L) in the formulation of autologous tumor cell-based vaccinesfor cancer patients with stage IV disease. Ann Surg Oncol 2007;14:869–84.

46. Heise C, Hermiston T, Johnson L, et al. An adenovirus E1A mutantthat demonstrates potent and selective systemic anti-tumoral effica-cy. Nat Med 2000;6:1134–9.

47. Alemany R. Cancer selective adenoviruses. Mol Aspects Med 2007;28:42–58.

48. Raki M, Sarkioja M, Desmond RA, et al. Oncolytic adenovirus Ad5/3-δ24 and chemotherapy for treatment of orthotopic ovarian cancer.Gynecol Oncol 2008;108:166–72.

49. Ghiringhelli F, Menard C, Puig PE, et al. Metronomic cyclophospha-mide regimen selectively depletes CD4+CD25+ regulatory T cellsand restores T and NK effector functions in end stage cancer pa-tients. Cancer Immunol Immunother 2007;56:641–8.

Cancer Res; 70(11) June 1, 2010 4309