rad001 (everolimus) improves the efficacy of replicating adenoviruses … · adenoviruses that...

10
RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses that Target Colon Cancer Krisztian Homicsko, Alexander Lukashev, and Richard D. Iggo National Center of Competence in Research Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC), Epalinges, Switzerland Abstract Selectively replicating adenoviruses have the potential to cure cancer but have shown little efficacy in clinical trials. We have tested the ability of the mTOR kinase inhibitor RAD001 (everolimus) to enhance the response of xenografts to an oncolytic adenovirus. The virus has Tcf sites inserted in the early viral promoters and replicates selectively in cells with activation of the Wnt signaling pathway. To enhance tumor cell infection, an integrin targeting peptide (CDCRGDCFC) was inserted into the fiber gene of the virus. RAD001 combines three useful properties: it inhibits tumor cell growth directly, blocks angiogenesis, and suppresses the immune response. RAD001 does not block viral protein expression, DNA replication, or cytopathic effect in tumor cells in vitro . After 6 weeks of daily RAD001 treatment, ongoing viral DNA replication could be detected in tumor xenografts, showing that RAD001 does not inhibit virus replication in vivo . I.v. injection of virus alone produced a small delay in xenograft growth, whereas combination therapy substantially prolonged the survival of the mice. We suggest that collapsing the tumor vasculature after the initial infection traps the virus and facilitates local spread within the tumor. Unlike conventional drugs, which require continued access to the tumor through the vascular system, oncolytic viruses are in principle less sensitive to late reductions in perfusion because they are produced locally within the tumor. (Cancer Res 2005; 65(15): 6882-90) Introduction Oncolytic viruses provide a means to target the causal oncogenic defects in tumors (1). Despite having great selectivity and efficacy in vitro , these viruses have shown only limited activity in preclinical xenograft models and in early stage clinical trials (2, 3). Many factors explain this low activity in vivo . The majority of i.v. administered virus is either sequestered on red cells or cleared by macrophages within minutes of injection (4, 5). This can be circumvented by depletion of Kupffer cells and coating the virus with hydrophilic polymers like hydroxypropyl methacrylamide (6), but delivery of the virus to the tumor in large amounts remains a major problem. Modification of the tropism of the virus by mutation of the fiber gene can reduce infection of nontumor cells, but it is difficult to achieve selectivity without loss of activity. The most successful strategies have instead sought to broaden the tropism to reduce the chance that tumor cells will be inefficiently infected because of poor expression of the natural receptors for human adenoviruses (7, 8). The CDCRGDCFC peptide was identified in a screen for homing of i.v.-injected phage to tumors (9). Tumor endothelial cells are known to express high levels of a receptor for RGD peptides, a v h 3 integrins, which explains the ability of RGD-modified phage to home to tumors (10). The RGD peptide binds to a wide range of integrins, some of which are highly expressed on tumors (11). Insertion of the CDCRGDCFC peptide into the HI loop of the fiber gene of adenovirus 5 (Ad5) has been shown to increase the efficiency of infection of many different cell types by recombinant adenoviruses (12). To improve the efficacy of oncolytic adenoviruses, they are routinely combined with chemotherapeutic drugs (13) or with prodrugs that can be activated by enzymes expressed by the virus (14). The drugs normally used to treat colon cancer are unattractive for combination therapy because they can inhibit viral replication: irinotecan inhibits topoisomerase I, 5-fluorouracil blocks nucleo- tide synthesis, and oxaliplatin cross-links DNA. Newer drugs offer the possibility to inhibit tumor cell growth without killing the virus. RAD001 (40-O -2-hydroxyethyl-rapamycin, everolimus) is an orally available derivative of the immunosuppressant macrolide rapamy- cin (15). Like rapamycin, RAD001 inhibits mTOR kinase, a key component of growth factor and nutrient signaling pathways (16, 17). Phosphorylation and consequent inactivation of 4E-BP1 by mTOR allows eIF4E to recruit eIF4F to the mRNA cap. This leads to enhanced translation of mRNAs with highly structured 5V -untranslated regions, including genes that promote cell growth and oncogenic transformation (18). RAD001 inhibits the growth of many cancer cell lines in vitro (19) and can inhibit the growth of tumor xenografts in vivo (20). One of the mechanisms of antitumor activity in vivo seems to be inhibition of angiogenesis (21), resulting in activity against xenografts of cell lines that respond only weakly to the drug in vitro . Mathematical modeling shows that the balance between the rate of tumor cell growth and virus spread is a critical determinant of the outcome of oncolytic virus infection (22). Whether it acts directly on tumor cells or indirectly by blocking tumor perfusion, the resulting delay in tumor growth induced by RAD001 should allow the virus to spread further within the tumor before the tumor cells can grow away from the foci of infection or the immune system halts the infection. The initial dosing regimens of RAD001 in cancer patients were chosen to avoid immunosuppression (23), but for combination therapy with virus, transient immunosuppression to extend the period of viral replication would be an advantage. Wnt pathway activation by mutations in the APC or b-catenin genes is seen in many different types of tumor, but is particularly common in colon cancer. We have previously developed oncolytic adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway (24, 25). These viruses have binding sites for the Tcf transcription factor in the early promoters. We describe here Tcf-regulated adenoviruses that have the peptide CDCRGDCFC inserted into the HI loop of the fiber gene to enhance Requests for reprints: Richard Iggo, Oncogene Group, Swiss Institute for Experimental Cancer Research, Ch. des Boveresses 155, CH-1066 Epalinges, Switzerland. Phone: 41-21-692-5889; Fax: 41-21-652-6933; E-mail: [email protected]. I2005 American Association for Cancer Research. Cancer Res 2005; 65: (15). August 1, 2005 6882 www.aacrjournals.org Research Article Research. on November 26, 2020. © 2005 American Association for Cancer cancerres.aacrjournals.org Downloaded from

Upload: others

Post on 17-Aug-2020

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

RAD001 (Everolimus) Improves the Efficacy of Replicating

Adenoviruses that Target Colon Cancer

Krisztian Homicsko, Alexander Lukashev, and Richard D. Iggo

National Center of Competence in Research Molecular Oncology, Swiss Institute for Experimental Cancer Research (ISREC),Epalinges, Switzerland

Abstract

Selectively replicating adenoviruses have the potential to curecancer but have shown little efficacy in clinical trials. We havetested the ability of the mTOR kinase inhibitor RAD001(everolimus) to enhance the response of xenografts to anoncolytic adenovirus. The virus has Tcf sites inserted in theearly viral promoters and replicates selectively in cells withactivation of the Wnt signaling pathway. To enhance tumorcell infection, an integrin targeting peptide (CDCRGDCFC)was inserted into the fiber gene of the virus. RAD001 combinesthree useful properties: it inhibits tumor cell growth directly,blocks angiogenesis, and suppresses the immune response.RAD001 does not block viral protein expression, DNAreplication, or cytopathic effect in tumor cells in vitro . After6 weeks of daily RAD001 treatment, ongoing viral DNAreplication could be detected in tumor xenografts, showingthat RAD001 does not inhibit virus replication in vivo . I.v.injection of virus alone produced a small delay in xenograftgrowth, whereas combination therapy substantially prolongedthe survival of the mice. We suggest that collapsing the tumorvasculature after the initial infection traps the virus andfacilitates local spread within the tumor. Unlike conventionaldrugs, which require continued access to the tumor through thevascular system, oncolytic viruses are in principle less sensitiveto late reductions in perfusion because they are producedlocally within the tumor. (Cancer Res 2005; 65(15): 6882-90)

Introduction

Oncolytic viruses provide a means to target the causal oncogenicdefects in tumors (1). Despite having great selectivity and efficacyin vitro , these viruses have shown only limited activity in preclinicalxenograft models and in early stage clinical trials (2, 3). Manyfactors explain this low activity in vivo . The majority of i.v.administered virus is either sequestered on red cells or cleared bymacrophages within minutes of injection (4, 5). This can becircumvented by depletion of Kupffer cells and coating the viruswith hydrophilic polymers like hydroxypropyl methacrylamide (6),but delivery of the virus to the tumor in large amounts remains amajor problem. Modification of the tropism of the virus bymutation of the fiber gene can reduce infection of nontumor cells,but it is difficult to achieve selectivity without loss of activity. Themost successful strategies have instead sought to broaden thetropism to reduce the chance that tumor cells will be inefficientlyinfected because of poor expression of the natural receptors for

human adenoviruses (7, 8). The CDCRGDCFC peptide wasidentified in a screen for homing of i.v.-injected phage to tumors(9). Tumor endothelial cells are known to express high levels of areceptor for RGD peptides, avh3 integrins, which explains theability of RGD-modified phage to home to tumors (10). The RGDpeptide binds to a wide range of integrins, some of which are highlyexpressed on tumors (11). Insertion of the CDCRGDCFC peptideinto the HI loop of the fiber gene of adenovirus 5 (Ad5) has beenshown to increase the efficiency of infection of many different celltypes by recombinant adenoviruses (12).To improve the efficacy of oncolytic adenoviruses, they are

routinely combined with chemotherapeutic drugs (13) or withprodrugs that can be activated by enzymes expressed by the virus(14). The drugs normally used to treat colon cancer are unattractivefor combination therapy because they can inhibit viral replication:irinotecan inhibits topoisomerase I, 5-fluorouracil blocks nucleo-tide synthesis, and oxaliplatin cross-links DNA. Newer drugs offerthe possibility to inhibit tumor cell growth without killing the virus.RAD001 (40-O-2-hydroxyethyl-rapamycin, everolimus) is an orallyavailable derivative of the immunosuppressant macrolide rapamy-cin (15). Like rapamycin, RAD001 inhibits mTOR kinase, a keycomponent of growth factor and nutrient signaling pathways(16, 17). Phosphorylation and consequent inactivation of 4E-BP1 bymTOR allows eIF4E to recruit eIF4F to the mRNA cap. Thisleads to enhanced translation of mRNAs with highly structured5V-untranslated regions, including genes that promote cell growthand oncogenic transformation (18). RAD001 inhibits the growth ofmany cancer cell lines in vitro (19) and can inhibit the growth oftumor xenografts in vivo (20). One of the mechanisms of antitumoractivity in vivo seems to be inhibition of angiogenesis (21), resultingin activity against xenografts of cell lines that respond only weaklyto the drug in vitro .Mathematical modeling shows that the balance between the rate

of tumor cell growth and virus spread is a critical determinant of theoutcome of oncolytic virus infection (22). Whether it acts directly ontumor cells or indirectly by blocking tumor perfusion, the resultingdelay in tumor growth induced by RAD001 should allow the virus tospread further within the tumor before the tumor cells can growaway from the foci of infection or the immune system halts theinfection. The initial dosing regimens of RAD001 in cancer patientswere chosen to avoid immunosuppression (23), but for combinationtherapy with virus, transient immunosuppression to extend theperiod of viral replication would be an advantage.Wnt pathway activation by mutations in the APC or b-catenin

genes is seen in many different types of tumor, but is particularlycommon in colon cancer. We have previously developed oncolyticadenoviruses that replicate selectively in cells with constitutiveactivation of the Wnt signaling pathway (24, 25). These viruses havebinding sites for the Tcf transcription factor in the early promoters.We describe here Tcf-regulated adenoviruses that have the peptideCDCRGDCFC inserted into the HI loop of the fiber gene to enhance

Requests for reprints: Richard Iggo, Oncogene Group, Swiss Institute forExperimental Cancer Research, Ch. des Boveresses 155, CH-1066 Epalinges,Switzerland. Phone: 41-21-692-5889; Fax: 41-21-652-6933; E-mail: [email protected].

I2005 American Association for Cancer Research.

Cancer Res 2005; 65: (15). August 1, 2005 6882 www.aacrjournals.org

Research Article

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 2: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

infection of tumor cells. We show that RAD001 does not inhibit thegrowth of these viruses in vitro or in vivo , and that combinationtherapy with RAD001 plus virus is much more effective in axenograft model than virus or RAD001 alone.

Materials and Methods

Chemicals. RAD001 (everolimus) was supplied by Novartis (Basel,

Switzerland) as dry powder for in vitro use and microemulsion for oral

use. The powder was dissolved in ethanol and stored as 10 mmol/L stock

solution at �20jC. The microemulsion (2% RAD001) and placebo emulsionwere aliquoted and stored at �20jC.

Cell lines. The colon cancer cell lines SW620, HT29, and Hct116 were

obtained from the American Type Culture Collection (ATCC, Manassas,

VA). LS174T and SW480 cells were provided by Dr. B. Sordat (ISREC,Epalinges, Switzerland). HER911 cells (26) were provided by Dr. P. Beard

(ISREC, Epalinges, Switzerland). HeLa cells were provided by Dr. D. Lane

(Department of Surgery, University of Dundee, Scotland). cR2 cells arederivatives of HEK293 cells that express a stable mutant of h-catenin, theviral polymerase and viral preterminal protein (27, 28). All cells were

cultured in DMEM (Invitrogen, Carlsbad, CA) with 10% fetal bovine

serum and 1% penicillin/streptomycin. RAD001 was used at finalconcentration of 3.6 Amol/L in vitro .

Viruses. The E1B-Tcf integrating vector, pRDI-241, was described by

Brunori et al. (25). The KpnI/XbaI Ad5 fragment (nucleotides 30,470 to

33,598) containing the fiber region was cloned from Ad5 genomic DNA(ATCC VR5) into pUC19 to give pCF159. The CDCRGDCFC motif was

inserted into the HI loop of the fiber by inverse PCR from pCF159 using

primers GGAGACTGTTTCTGCCCAAGTGCATACTCTATGTC (oKH11) andGCGGCAGTCACAAGTTGTGTCTCCTGTTTCCT (oKH12) to create pKH67.

The Coxsackie-adenovirus receptor (CAR) binding site was mutated in the

knob of the fiber by inverse PCR from pKH67 using primers GGTGGTGGA-

GATGCTAAACTCACTTTGGTC (oKH9) and ATTTAGACTACAGTTAGGA-GATGGAGCTGG (oKH10) to create pKH68. The amino acid sequence of the

modified fiber around the mutation is NCRLNAEKDAK in the wild-type and

NCSLNGGGDAK in the mutant (29). The KpnI/XbaI fragments of pKH67

and pKH68 were cloned into pRS406 (30) to create pKH69 (RGD insertion)and pKH70 (RGD insertion and CAR deletion), the mutant fiber integrating

vectors. The adenovirus genome was modified by two-step gene

replacement as described by Gagnebin et al. (31). First, the Tcf-E1B sites

were inserted into vpCF11 (Tcf-E1A and Tcf-E4; ref. 24) using pRDI-241 tocreate vpKH1. Subsequently, the fiber mutations were inserted in vpKH1

using pKH69 and pKH70 to create vpKH3 and vpKH6, respectively. The

vKH1, vKH3, and vKH6 viruses were produced by transfection of PacI-digested vpKH1, vpKH3, and vpKH6 into cR2 cells (27). The viruses were

then plaque purified on SW480 cells, expanded on SW480 cells using Cell

Factories (Nunc, Roskilde, Denmark), purified by double CsCl banding,

buffer exchanged using HR400 columns (Amersham, Little Chalfont, UnitedKingdom) into 1 mol/L NaCl, 100 mmol/L Tris-HCl (pH 8.0), 10% glycerol,

and stored frozen at �70jC. The identity of each batch was checked by

restriction digestion and automated fluorescent sequencing in the E1B

(nucleotides 1,300 to 2,300) and fiber (nucleotides 30,470-33,598) regionsusing the following primers: E1B sense, TGTCTGAACCTGAGCCTGAG; fiber

antisense, CTACTGTAATGGCACCTG; fiber sense, GCCATTAATGCAGGA-

GATG. Apart from the desired mutations, no differences were foundbetween the sequences of VR5 and the Tcf viruses. Particle counts were

based on the absorbance at 260 nm (A260) of virus in 0.1% SDS, using the

formula 1 A260 = 1012 particles/mL.

Western blotting. Cells were infected with 100 particles/cell in DMEM.Two hours after infection, the medium was replaced with complete

medium. Cells were harvested at the indicated times in SDS-PAGE sample

buffer. E1A, DBP, and fiber were detected with the M73 (Santa Cruz

Biotechnology, Santa Cruz, CA), B6 (32), and 53 4D2 (Research Diagnostics,Inc., Flanders, NJ) antibodies, respectively.

Cytopathic effect assay. Cells in six-well plates were infectedwith 10-folddilutions of virus in DMEM. Four hours after infection, the medium wasreplaced with complete medium containing RAD001. Four days after infec-

tion, fresh medium containing RAD001 was added. After 5 to 8 days, the cellswere fixed with 4% formaldehyde in PBS and stained with crystal violet.

Quantitative PCR assay. Cells were infected in vitro with one viral

particle per cell in DMEM. The medium was replaced after 4 hours and

cells were harvested after 48 hours. DNA was purified from tissue culturecells and mouse tissues using a DNeasy kit (Qiagen, Hilden, Germany)

according to the manufacturer’s instructions. Quantitative PCR was done

on a Light Cycler (Roche, Basel, Switzerland) using FastStart SYBR Green

(Roche), 500 nmol/L primers (Invitrogen), and 10 ng template DNA.Results were expressed relative to cell number assuming 6 pg DNA/cell.

The primers used for quantitative PCR were hexon forward primer,

CTTCGATGATGCCGCAGTG and hexon reverse primer, GGGCTCAGGTA-

CTCCGAGG.Animal experiments. Four-week-old female NMRI nu/nu mice were

purchased from Elevage Janvier (Le Genest St Isle, France). S.c. SW620 flank

xenografts were made by injecting 107 cells. Mice ( five per group) were

injected with virus when tumors reached 80 to 150 mm3 in size. A total of

1 � 1011 particles were injected into the tail vein per day, given as four shots

of 2.5 � 1010 particles at four hourly intervals. Two regimens were tested:

virus injection on a single day at the start of the experiment (total dose per

mouse 1 � 1011 particles) or virus injection at weekly intervals for 3 weeks

(total dose 3 � 1011 particles). RAD001 microemulsion was diluted in water

and 5 mg/kg/d was administered by daily gavage starting the day after virus

injection. For mice receiving weekly injections of virus, RAD001 was omitted

the day before and on the day of the injection; RAD001 administration

continued daily thereafter to the end of the experiment. Tumor size was

measured every 2 days. Tumor volume was calculated according to the

formula, volume = length � width2 � 3.14/6. Differences in growth curves

were calculated by two-tailed Student’s t test. Differences in survival curves

were calculated by log-rank test in R (33).

ELISA. Three weeks after i.v. injection of 109 viral particles of adenovirus

(vKH1), 200 AL of blood was taken from each mouse. The serum titer of

antibodies against adenoviral proteins was measured by ELISA using ELISA

plates (Nunc) coated with Ad5 as described (34).Fluorescence in situ hybridization. Viral genomic DNA was labeled

using the DIG DNA labeling kit (Roche). Tumor sections were fixed in

4% PFA, digested for 30 minutes with proteinase-K, acetylated with TEAfor 10 minutes, and blocked with hybridization buffer without probe for

2 hours. Labeled probe was added and slides were hybridized overnight at

50jC. After washing in SSC, slides were incubated overnight with anti-DIG

horseradish peroxidase at 4jC and label was detected with TSA Cy3 reagentkit (Perkin-Elmer, Boston, MA). 4V,6-Diamidino-2-phenylindole (DAPI) was

used to counterstain nuclei.

Results

The structure of the viruses used in this study is shown inFig. 1A . The parental virus, vKH1, has Tcf sites inserted into theinverted terminal repeats and E1B promoter, resulting in Tcf-dependent expression of E1A, E1B and, to a lesser extent, E4 (24).The peptide CDCRGDCFC was inserted into the HI loop of thefiber gene to confer on vKH6 the ability to infect cells eitherthrough binding to the normal receptor (CAR) or through bindingto integrins, particularly avh3 and avh5. The CAR binding site inthe fiber gene of vKH6 was deleted to create vKH3. To avoidselection for escape from the Tcf-based regulation of the earlypromoters, the viruses were produced in SW480 cells, which haveconstitutively high Tcf activity because of a mutation in the APCgene (35). The particle to plaque-forming unit (pfu) ratio of theviruses is shown in Fig. 1A , based on infection of HER911 cells,which are permissive for the Tcf viruses because of trans-complementation of the regulated promoters. The particle to pfuratio was much higher for vKH3 than for the other viruses,indicating that CAR plays a major role in HER911 infection. Toavoid infecting cells with units of virus defined according to

Combination Therapy with Oncolytic Virus and RAD001

www.aacrjournals.org 6883 Cancer Res 2005; 65: (15). August 1, 2005

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 3: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

misleading criteria, particle counts rather than pfu were usedto calculate the viral titer for the experiments described below.Western blotting for the E1A, DBP, and fiber proteins was done

to test whether the new Tcf viruses are able to infect colon cancercells efficiently and retain selectivity. We tested three colon cancercell lines with strong Tcf activity, SW480, SW620, and LS174T, andtwo colon cancer cell lines with weak Tcf activity, HT29 and Hct116(35). A cervical cancer cell line without activation of the Wntpathway, HeLa, was used as a negative control. Viral proteinexpression by the Tcf viruses was reduced or absent in HeLa cells,confirming our previous results with Tcf-regulated viruses andshowing that fiber modification does not compromise promoterselectivity (Fig. 1B). The RGD viruses were at least as good as vKH1in all of the colon cancer cell lines, and gave substantially higherviral protein expression in LS174T, indicating that integrin bindingplays an important role in infection of LS174T cells.To determine whether addition of the RGD peptide changes the

in vitro efficacy of the viruses, cytopathic effect assays were done(Fig. 1C). At low multiplicities of infection, virus must undergo

multiple rounds of productive infection to kill all the cells. Thus,the assay measures the ability of the virus both to infect cells andto replicate. Insertion of the RGD peptide into the fiber increasedthe efficacy of the Tcf-regulated viruses on colon cells by a factor of10 to 100. vKH3, in which the CAR-binding site in the fiber has beendeleted, showed intermediate cytopathic effect in LS174T andHct116, indicating that both CAR and integrins are used by vKH6to infect these cells. The efficacy of vKH6 was comparable with, orgreater than, that of wild-type Ad5 in all of the colon cancer celllines tested except HT29, which has the lowest Tcf activity. vKH1and vKH3 were 10,000-fold less cytopathic than wild-type virus onthe HeLa cells. vKH6 was 10-fold more cytopathic than the otherTcf-regulated viruses on HeLa cells, showing that integrins andCAR both contribute to HeLa infection. The perfect control forvKH6 would be a virus with wild-type promoters and RGDinsertion in the fiber, but this virus was not constructed forbiosafety reasons. The increase in cytopathic effect of vKH6 onHeLa cells probably reflects a large increase in the effectivemultiplicity of infection (number of viral genomes entering the cell)

Figure 1. In vitro characterization of Tcf-regulated adenoviruses. A, adenoviruses used in this study. Part/pfu, the ratio of particles measured by A260 to pfumeasured on HER911 cells. The multiplicity of infection (moi) is based on particles/cell for all experiments described. B, Western blot for E1A, DBP, and fiber proteinexpression. SW620, SW480, HT29, Hct116, and LS174T cells were infected at a moi of 100. HeLa cells were infected at a moi of 1,000. Samples were collectedat the indicated time points. C, cytopathic effect assay on colon cancer cell lines (SW620, SW480, LS174T, HT29, and Hct116) and HeLa cells. Cultures were infectedwith 10-fold dilutions of virus starting at a moi of 1,000, and stained with crystal violet on day 6 for SW620 and day 8 for the rest.

Cancer Research

Cancer Res 2005; 65: (15). August 1, 2005 6884 www.aacrjournals.org

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 4: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

caused by the RGD modification, rather than any change in the Tcfregulation of the viral promoters. We conclude that the viruses withmodified fiber genes retain selectivity for colon cancer cells andshow similar or greater cytopathic effect than the parental virus.This is consistent with previous reports on the behavior of RGD-modified adenoviruses (36).To determine whether addition of the RGD peptide alters the

pattern of Tcf virus infection in vivo , the virus was injected into thetail vein of nude mice with s.c. SW620 xenografts. The virus wasinjected in four aliquots at 4 hourly intervals. This schedule wasused to prolong the circulation time of the virus in the blood andhence the probability of infection (37): The first dose is rapidlycleared by Kupffer cells, which are themselves eliminated in theprocess. Fluorescence in situ hybridization (FISH) to viral DNA wasused to identify sites of virus replication; single viral genomes werebelow the detection limit of the assay. Nine days after injection,vKH1 produced compact islands of infected cells, whereas vKH6produced a more diffuse pattern, suggesting that vKH6 may spreadbetter through tumor tissue (Fig. 2A). To determine whether thespectrum of organs infected was altered by insertion of the RGDpeptide, which can bind to multiple different integrins, thebiodistribution was tested (Fig. 2B). Virus was injected into thetail vein as described above and organs were harvested forquantitative PCR 24 hours later. Compared to other organs, therewas less viral DNA than expected in liver after injection of the viruswith native tropism (vKH1). This may be related to the fractionateddosing schedule, which leads to clearance of Kupffer cells. Theamount of viral DNA was 10 to 100 times higher in the tumorsinfected with the RGD viruses, consistent with the in vitro datashowing that SW620 is more susceptible to infection with vKH6than vKH1. The amount of viral DNA was similar for the viruseswith native or modified tropism in all mouse organs except liver,where the amount was increased 50-fold for vKH3 and vKH6. Wild-type adenovirus is lethal after i.v. injection of 1010 particles(Fig. 2C). This is caused by virus replication in the liver andfulminant hepatocyte necrosis (38). None of the mice treated withthe Tcf viruses developed fatal liver necrosis after injection of 10times the lethal dose of wild-type virus (Fig. 2C). Consistent withthe increased liver infection seen by quantitation of viral DNA,there was an inflammatory infiltrate in the region around thecentral veins of the mice 3 days after receiving vKH6 (Fig. 2C ,arrows). The inflammatory infiltrates resolved completely and wereundetectable 20 days after the last virus injection (data not shown).The lack of overt signs of illness in the mice receiving the Tcfviruses indicates that the increased ability of the RGD viruses toinfect the liver is more than offset by the reduction in toxicitycaused by Tcf regulation of the early promoters.To test whether the increased ability of vKH6 to infect SW620

cells in vitro leads to increased efficacy in vivo , xenograft growthwas analyzed after i.v. injection of virus. Two regimens werecompared: injection on a single day (total 1011 particles injected)and injection on 3 days at weekly intervals (total 3 � 1011 particlesinjected). In both cases, the virus was fractionated on the dayof injection to circumvent Kupffer cell clearance. Three courses ofinjection at weekly intervals (Fig. 3A , open symbols) were moreeffective than injection on a single day (closed symbols). The RGD-modified virus, vKH6 (triangles), was more effective than vKH1(circles). Even in the best case, however, most of the tumorseventually relapsed. Quantitative PCR showed that there was a largeamount of viral DNA present in the tumors at the time of relapse(data not shown). To understand why the virus had failed to control

tumor growth, tumors were examined for viral DNA by FISH atdifferent time points. Three days after infection, virus was found inisolated clusters of cells, corresponding to the cells infected with theinjected virus and its progeny after one cycle of replication. At later

Figure 2. In vivo characterization of Tcf-regulated adenoviruses. A, FISH for viralDNA in SW620 xenografts 5 days after i.v. injection of 1011 particles of vKH1 andvKH6. Red, viral DNA (FISH); blue, cell nuclei (DAPI). Bar, 200 Am. B,biodistribution of vKH1, vKH3, and vKH6 24 hours after i.v. injection of 1011

particles of virus. Each column represents three individual experiments. Viral DNAwas measured by quantitative PCR. C, liver histology 3 days after i.v. injection ofvirus. Wild-type Ad5, 1010 particles; vKH1 and vKH6, 1011 particles. Arrows showinflammatory infiltrates around the central vein with vKH6. Bar, 100 Am.

Combination Therapy with Oncolytic Virus and RAD001

www.aacrjournals.org 6885 Cancer Res 2005; 65: (15). August 1, 2005

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 5: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

time points, the infected regions expanded to form confluent areasof infection in some parts of the tumor (Fig. 3B). Relapse can beexplained by uncontrolled growth of tumor regions that are devoidof virus. This is consistent with theoretical models showing thattumor cell spread generally outpaces virus spread (22).RAD001 is an mTOR inhibitor that inhibits tumor cell growth

directly, blocks angiogenesis, and suppresses the immune response.To determine whether RAD001 interferes with translation ofadenoviral proteins, a Western blot was done in the presence orabsence of RAD001. There was no evidence of inhibition of viralprotein expression (Fig. 4A). Quantitation of viral DNA 48 hoursafter infection showed only a small effect of RAD001 (in most celllines, there was a slight increase in replication in the presence ofthe drug; Fig. 4B). In vitro efficacy was tested in cytopathic effectand 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromideassays (Fig. 4C ; data not shown). Both showed either no effect ora 5- to 10-fold increase in efficacy in the presence of the drug. Totest whether RAD001 inhibits replication in vivo , an SW620xenograft was infected by the i.v. route as above, and the amountand distribution of viral DNA in the tumor were examined after

prolonged treatment of the mice with RAD001. Quantitative PCRshowed that a large amount of viral DNA was present in tumorseven after treatment with RAD001 for 6 weeks (Fig. 5A ).Widespread areas of the tumor were also positive by FISH forviral DNA after RAD001 treatment for 6 weeks (Fig. 5B). Some ofthe FISH signal comes from cell debris in areas of necrosis, but thestrongest signals are from the nuclei of living cells that areundergoing productive viral infection. We conclude that RAD001does not inhibit virus growth in tumor cells in vitro or in vivo . Inaddition to its effect on mTOR in the tumor, RAD001 is known tosuppress the immune response by inhibiting mTOR in lympho-cytes. The induction of a neutralizing antibody response is thoughtto limit the efficacy of oncolytic adenoviruses in vivo (39). Toconfirm that RAD001 can prevent induction of a primary antibodyresponse against adenovirus, immunocompetent mice were treatedwith RAD001 and then immunized with adenovirus. As expected,RAD001 was able completely to block the primary antibodyresponse to virus (Fig. 5C).

Figure 4. Effect of RAD001 on vKH6 virus infection in vitro . A, Western blot forE1A and DBP protein expression 24 and 48 hours after infection. B, viral DNAreplication was analyzed by quantitative PCR for viral DNA 48 hours afterinfection. Cells were infected at a moi of 1. RAD001 was added 4 hours afterinfection. C, cytopathic effect assay in the presence (+) or absence (�) ofRAD001. Cells were infected with 10-fold dilutions of virus starting at a moi of1,000 and stained with crystal violet on day 6.

Figure 3. Xenograft infection with virus alone. A, growth curves of s.c.SW620 xenografts in nude mice (five mice per group). Viral treatment startedwhen tumors reached f100 mm3. Circles , vKH1. Triangles, vKH6. Closedsymbols, 1011 particles of virus i.v. on day 0. Open symbols, 1011 particles ofvirus i.v. on days 0, 7, and 14. �, control (buffer injection). B, FISH for viralDNA in tumors 9 days after i.v. injection of 1011 particles of virus. The wholetumor is shown to demonstrate the patchy distribution of virus. Red, viral DNA(FISH); blue, cell nuclei (DAPI). Bar, 500 Am. The red fluorescence at thetumor margin (arrow) is caused by autofluorescence of erythrocytes nothybridization to viral DNA.

Cancer Research

Cancer Res 2005; 65: (15). August 1, 2005 6886 www.aacrjournals.org

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 6: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

The efficacy of combination therapy in vivo was tested by givingi.v. virus followed by daily oral RAD001. The virus was givenaccording to the same schedules as in Fig. 3A . RAD001 was withheldthe day before and on the day of virus injection, on the grounds thatantivascular effects of the drug might limit access of the virus to thetumor. Mice treated with placebo (gavage with the vehicle used forthe RAD001) all had to be sacrificed because of uncontrolled tumorgrowth by day 20 (Fig. 6A). This reflects the rapid growth of SW620xenografts once the tumors reach f100 mm3. The start of theexperiment was delayed until the tumors reached 80 to 150 mm3 toensure that they had developed a vascular tree that could be calledon to deliver the virus after i.v. injection. Consistent with the knownlow toxicity of RAD001, there was no weight loss in RAD001-treatedmice after 20 days of treatment. RAD001 alone was more effectivethan virus alone in slowing growth of the tumors, but by 40 daysmost mice still had to be sacrificed because of progressive tumorgrowth (Fig. 6B). Tumors treated with the combination showedmarkedly better responses: Only one mouse in each vKH1 plusRAD001 group had to be sacrificed, and no mice in either vKH6 plusRAD001 group had to be sacrificed before the end of the experiment40 days after the first virus injection (Fig. 6B). The differencebetween the survival of the control and virus + RAD001 groups wassignificant in every case (P < 0.01). The smallest difference insurvival was between that of the RAD001 alone and virus + RAD001groups: It achieved significance (P < 0.05) for the 1� vKH1, 1�vKH6, and 3� vKH6 groups but not the 3� vKH1 group. The failureof the 3�vKH1 + RAD001 versus RAD001 alone survival differenceto reach significance is explained by the small number of animals.Analysis of the growth curves (Fig. 6A) showed that at the time thefirst mouse was sacrificed in the RAD001 alone group (day 28), therewas a significant difference in tumor size in the 3� vKH1 + RAD001versus RAD001 alone groups (P < 0.05). The mechanism for theinteraction between oncolytic virus and RAD001 is unlikely to berelated to the immunosuppressant effect of the drug because themice were immunodeficient to allow xenografting. It could bepartially explained by a direct inhibitory effect on the tumor cells,given that the drug slightly increased the cytopathic effect of thevirus in vitro (Fig. 4C). Histologic examination of the tumors showedthat RAD001-treated tumors had the typical appearance of poorlyvascularized tumors, with a rim of viable cells and a large centralnecrotic core, in contrast with the untreated tumors, whichcontained uniform sheets of viable cells with only patchy necrosis(Fig. 5D). It is likely that this antivascular effect contributedimportantly to the interaction between virus and drug, substantiallydelaying the growth of the tumor while still allowing the virus tospread within it.

Discussion

The major current limitation to the development of oncolyticviruses is the low efficacy of the viruses in vivo . This is the firstreport showing systemic efficacy of Tcf-regulated adenoviruses inmice. Virus plus RAD001 significantly slowed tumor growth andprolonged the survival of the treated animals. Combination therapywith RAD001 and RGD-modified viruses gave the best results.Although the ideal targeting ligand would lead to selective

infection of cells in the tumor, no viruses selectively infectingtumor cells have yet shown high efficacy in vivo (7). The mainconcern is to preserve activity of the virus on tumor cells. The twoapproaches most widely used are to target heparan sulfateproteoglycans with poly-lysine or to target integrins with RGD.

Figure 5. Effect of RAD001 in vivo . A, quantitative PCR for viral DNA intumors 6 weeks after i.v. injection of vKH1 or vKH6. The mice received 1 or3 days of treatment with 1011 particles of virus per day and daily RAD001.B, FISH for viral DNA in a tumor 6 weeks after i.v. injection of 1011 particles ofvKH1 and daily RAD001. Red, viral DNA (FISH); blue, cell nuclei (DAPI). Bar,100 Am. C, ELISA for antiadenovirus antibody. Groups of four mice receivedRAD001 or placebo for 5 days followed by i.v. injection of 109 particles ofvKH1. The titer of antiadenovirus antibodies in serum was measured by ELISA21 days after infection. D, histology of s.c. SW620 xenografts in mice treatedwith RAD001. Left, day 20 after i.v. injection of vKH1 1011 particles; right, day40 after i.v. injection of vKH1 1011 particles and daily RAD001. H&E staining.Bar, 100 Am.

Combination Therapy with Oncolytic Virus and RAD001

www.aacrjournals.org 6887 Cancer Res 2005; 65: (15). August 1, 2005

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 7: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

Neither change impairs the ability of the virus to infect cellsthrough CAR. A virus using all three routes of infection has beenproduced and shows promise in ovarian cancer (8). A potentialdrawback of this approach is that the viruses are not tumorspecific, and side effects caused by infection of normal cells arepotentially increased. Insertion of the RGD peptide into the Tcf-regulated viruses substantially enhanced its ability to infect tumorcells, and increased the amount of viral DNA that could berecovered from tumor and liver at 24 hours. The normal tissues

where the Wnt pathway is likely to be active are stem cells in theskin, hematopoietic system and intestine, and neurons in theseveral regions of the brain, including the subventricular zone,cortex, and hippocampus (40–42). There was a small increase inthe amount of viral DNA detectable in skin with vKH6, albeit to avery low level, and no change in the amount of viral DNA in brainor intestine. The liver was the major site of toxicity. The efficiencyof infection depends on the relative expression of receptors on thecells and ligands on the viruses. It is likely that insertion of the RGD

Figure 6. Effect of RAD001 on tumor response in vivo . A, growth curves of s.c. SW620 xenografts in NMRI nu/nu mice. E, vKH6 and daily RAD001; ., vKH1and daily RAD001; n, daily RAD001 alone; �, control. Virus injections are indicated by arrows. 1� virus injection: mice received 1011 particles of the virus i.v. on day 0;3� virus injection: mice received 1011 particles of the virus i.v. on days 0, 7, and 14. The control group received placebo (RAD001 vehicle) by daily gavage.B, Kaplan-Meier curves showing the fraction of mice with SW620 xenografts <1,000 mm3 [the corresponding growth curves are shown in Figs. 3A and (A )]. Eachgroup contained five mice. Virus injections are indicated by arrows. V+R, virus plus RAD001; R, RAD001 alone; V, virus alone; C, control.

Cancer Research

Cancer Res 2005; 65: (15). August 1, 2005 6888 www.aacrjournals.org

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 8: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

peptide alters the distribution of virus within the liver to takeadvantage of integrin expression by hepatocytes (43). Liverinfection resulted in the formation of inflammatory infiltrates atearly time points. In immunocompetent mice, the acute response isnormally followed by T-cell infiltration and acquired immunity. Inthe immunodeficient mice used here, the inflammatory infiltratescompletely resolved by 20 days. Inflammatory infiltrates around thecentral veins in the liver were accompanied by viral replication inoccasional hepatocytes, which could be detected by FISH.1

Compared with infection by wild-type virus, which producedfulminant liver necrosis after injection of 1010 particles, the RGDviruses were very well tolerated by the mice after injection of 10times more virus. This must reflect the decrease in expression ofviral proteins caused by Tcf regulation of the early promoters. Wehave not constructed Ad5 with wild-type promoters and RGD inthe fiber, but one might reasonably expect such a virus to produceliver necrosis at lower doses than the wild-type virus. Compared tosuch a virus, our Tcf-regulated viruses would probably be >100-foldless toxic to the liver. The other major toxicity that has beenreported for systemically administered adenoviral vectors in miceis acute hypotension (44). About one third of the mice in the vKH6group showed signs consistent with hypotension after the firstinjection of virus, but no mice died as a result. There were no overtclinical changes after subsequent injections, consistent withreports that the initial hypotension is caused by activation ofKupffer cells, which are then eliminated (44).Many classic chemotherapeutic drugs have been tested in

combination with oncolytic viruses (13). With the exception oftaxanes, which act on microtubules, most of these drugs areexpected to inhibit viral DNA replication. RAD001 has severalattractive properties for combination therapy with oncolyticviruses. It has no known inhibitory effect on viral DNA replication,it inhibits tumor cell growth directly, it blocks tumor angiogenesis,and it specifically inhibits the immune response. A majoradvantage of RAD001 is its low toxicity compared with standardchemotherapeutic agents. RAD001 inhibits translation of cellularmRNAs with highly structured 5V-untranslated regions by blockingphosphorylation of 4E-BP1 (18). Early in adenovirus infections,there is an E1A-dependent increase in 4E-BP1 phosphorylation,which can be blocked by rapamycin (45). Despite this potentiallyadverse interaction, there was a small increase in cytopathic effectand viral DNA replication in the presence of RAD001 in most of thetumor cell lines we tested. One interpretation is that induction of4E-BP1 phosphorylation by the virus is more important inquiescent normal cells than in tumor cells. Late in infection, thevirus uses the L4 100 K protein to inhibit translation of cappedcellular mRNAs by competing for Mnk1 binding to eIF4G (46). The100 K protein contains an RNA-binding domain that binds to thetripartite leader sequence in late mRNAs. This allows selectivetranslation of late viral RNAs by ribosome shunting, a process thatis independent of scanning by eIF4A (47). We speculate that thisprocess is largely resistant to RAD001.RAD001 is an orally active derivative of rapamycin, a well-

characterized immunosuppressant that is used clinically in organtransplant recipients. Therapy with oncolytic adenoviruses inducesa strong neutralizing antibody response in all patients (48). This isa particular concern for treatment schedules involving multiplecycles of virus injection. Most viruses already manipulate the

immune system in some way, but total suppression of the immuneresponse by a virally encoded gene would raise obvious biosafetyconcerns. Transient immune suppression by a drug avoids thisproblem. Several approaches have been tested to suppress theimmune response, including the use of anti-CD40 antibodies (49).RAD001 is more suitable for combination with viral therapy thancyclosporin, because cyclosporin stimulates the formation of tumorblood vessels (21). We have shown that RAD001 can suppress theprimary immune response to adenovirus in mice (Fig. 5C), butmost patients already have anti-Ad5 antibodies following Ad5infection in childhood. This means that what is required is theability to suppress a secondary immune response, which is a muchmore difficult task. One way to investigate this problem would beto use immunocompetent mouse tumor models, some of whichmay be permissive for our Tcf-regulated viruses (50).We have shown that combination therapy with oncolytic viruses

and RAD001 is more effective than treatment with either agentalone. Mathematical modeling suggests that seeding of virus tomultiple sites in the tumor is essential to achieve rapid spread of thevirus throughout the tumor mass, which is a prerequisite for cure ofthe disease (22). The main requirement is to deliver large amountsof virus to as many regions of the tumor as possible at the start oftreatment. Tumor blood vessels are disorganized and leaky, whichshould favor escape of the virus from the circulation, but this iscounterbalanced by high interstitial fluid pressure within the tumorand a thickened basement membrane, which are known to reducedelivery of macromolecules to the tumor (51). Antiangiogenictherapy, for example, with antibodies against vascular endothelialgrowth factor (VEGF, bevacizumab) or VEGFR2 (CD101), transientlynormalizes the vasculature and lowers interstitial fluid pressure(52, 53). This so-called ‘‘normalization window’’ is associated withpericyte recruitment and thinning of the basement membrane.Adenovirus is large (90 nm) compared with normal drugs, butsmaller than the pores in tumor blood vessels (>200 nm; ref. 54). Theideal solution would be to normalize the vessels to thin thebasement membrane and then transiently increase vessel perme-ability at the time of virus injection. TNFa is known to increase thepermeability of tumor blood vessels, but in preliminary experimentswe observed increased hepatotoxicity after low-dose TNFa therapy.Systemic therapy with VEGF is unattractive because it increasespermeability indiscriminately (55). Rapamycin and RAD001 blockVEGF induction (21).2 We tried to exploit this fact in our weeklydosing schedule by withdrawing RAD001 the day before givingthe virus. This schedule might result in normalization of the vascu-lature during RAD001 treatment, followed by induction of vesselleakiness by a burst of VEGF secretion around the time of virusinjection. Because the precise timing of the change in perfusionupon RAD001 treatment is unclear, it is possible that otherschedules would give even better results. There are clearly manypossible levels of interaction between antiangiogenic therapy andoncolytic viruses. The important conclusion from our study is thatoncolytic viruses are particularly well suited to combination withantiangiogenic therapy.

Acknowledgments

Received 1/29/2005; revised 3/28/2005; accepted 5/22/2005.Grant support: Swiss National Science Foundation NCCR Molecular Oncology

Program (K. Homicsko, A. Lukashev, and R. Iggo).

1 K. Homicsko and R. Iggo, unpublished data. 2 H.A. Lane, personal communication.

Combination Therapy with Oncolytic Virus and RAD001

www.aacrjournals.org 6889 Cancer Res 2005; 65: (15). August 1, 2005

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 9: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

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

We thank Dr. H.A. Lane (Novartis, Basel, Switzerland) for providing RAD001, theSwiss Institute for Experimental Cancer Research microscopy facility for technicalassistance, and Drs. H.A. Lane and P. Beard for critical reading of the manuscript.

References

1. Hawkins LK, Lemoine NR, Kirn D. Oncolytic bio-therapy: a novel therapeutic platform. Lancet Oncol2002;3:17–26.

2. Kirn D. Oncolytic virotherapy for cancer with theadenovirus dl1520 (Onyx-015): results of phase I and IItrials. Expert Opin Biol Ther 2001;1:525–38.

3. Reid T, Warren R, Kirn D. Intravascular adenoviralagents in cancer patients: lessons from clinical trials.Cancer Gene Ther 2002;9:979–86.

4. Alemany R, Suzuki K, Curiel DT. Blood clearance ratesof adenovirus type 5 in mice. J Gen Virol 2000;81:2605–9.

5. Cichon G, Boeckh-Herwig S, Kuemin D, et al. Titerdetermination of Ad5 in blood: a cautionary note. GeneTher 2003;10:1012–7.

6. Green NK, Herbert CW, Hale SJ, et al. Extended plasmacirculation time and decreased toxicity of polymer-coated adenovirus. Gene Ther 2004;11:1256–63.

7. Wu H, Seki T, Dmitriev I, et al. Double modifica-tion of adenovirus fiber with RGD and polylysinemotifs improves coxsackievirus-adenovirus receptor-independent gene transfer efficiency. Hum Gene Ther2002;13:1647–53.

8. Wu H, Han T, Lam JT, et al. Preclinical evaluation of aclass of infectivity-enhanced adenoviral vectors inovarian cancer gene therapy. Gene Ther 2004;11:874–8.

9. Pasqualini R, Koivunen E, Ruoslahti E. av integrins asreceptors for tumor targeting by circulating ligands. NatBiotechnol 1997;15:542–6.

10. Ruoslahti E, Rajotte D. An address system in thevasculature of normal tissues and tumors. Annu RevImmunol 2000;18:813–27.

11. Ruoslahti E. RGD and other recognition sequencesfor integrins. Annu Rev Cell Dev Biol 1996;12:697–715.

12. Dmitriev I, Krasnykh V, Miller CR, et al. Anadenovirus vector with genetically modified fibersdemonstrates expanded tropism via utilization of acoxsackievirus and adenovirus receptor-independentcell entry mechanism. J Virol 1998;72:9706–13.

13. Heise C, Sampson-Johannes A, Williams A,McCormick F, Von Hoff DD, Kirn DH. ONYX-015, anE1B gene-attenuated adenovirus, causes tumor-specificcytolysis and antitumoral efficacy that can be augmentedby standard chemotherapeutic agents. Nat Med 1997;3:639–45.

14. Kirn D, Niculescu-Duvaz I, Hallden G, Springer CJ.The emerging fields of suicide gene therapy andvirotherapy. Trends Mol Med 2002;8:S68–73.

15. Huang S, Houghton PJ. Targeting mTOR signaling forcancer therapy. Curr Opin Pharmacol 2003;3:371–7.

16. Jacinto E, Hall MN. Tor signalling in bugs, brain andbrawn. Nat Rev Mol Cell Biol 2003;4:117–26.

17. Shamji AF, Nghiem P, Schreiber SL. Integration ofgrowth factor and nutrient signaling: implications forcancer biology. Mol Cell 2003;12:271–80.

18. Mamane Y, Petroulakis E, Rong L, Yoshida K, Ler LW,Sonenberg N. eIF4E—from translation to transforma-tion. Oncogene 2004;23:3172–9.

19. Beuvink I, O’Relly T, Zumstein S, et al. Antitumoractivity of RAD-001, an orally active rapamycin deriva-tive. Proc Am Assoc Cancer Res 2001;42:1972.

20. Boulay A, Zumstein-Mecker S, Stephan C, et al.Antitumor efficacy of intermittent treatment scheduleswith the rapamycin derivative RAD001 correlates withprolonged inactivation of ribosomal protein S6 kinase 1in peripheral blood mononuclear cells. Cancer Res 2004;64:252–61.

21. Guba M, von Breitenbuch P, Steinbauer M, et al.Rapamycin inhibits primary and metastatic tumorgrowth by antiangiogenesis: involvement of vascularendothelial growth factor. Nat Med 2002;8:128–35.

22. Wein LM, Wu JT, Kirn DH. Validation and analysis ofa mathematical model of a replication-competentoncolytic virus for cancer treatment: implications forvirus design and delivery. Cancer Res 2003;63:1317–24.

23. Van Oosterom AT, Dumez H, Desai J, et al. Combina-tion signal transduction inhibition: a phase I/II trial ofthe oral mTOR-inhibitor everolimus (E, RAD001) andimatinib mesylate (IM) in patients (pts) with gastroin-testinal stromal tumor (GIST) refractory to IM. J ClinOncol 2004;22:3002.

24. Fuerer C, Iggo R. Adenoviruses with Tcf binding sitesin multiple early promoters show enhanced selectivityfor tumour cells with constitutive activation of the wntsignalling pathway. Gene Ther 2002;9:270–81.

25. Brunori M, Malerba M, Kashiwazaki H, Iggo R.Replicating adenoviruses that target tumors withconstitutive activation of the wnt signaling pathway.J Virol 2001;75:2857–65.

26. Fallaux FJ, Kranenburg O, Cramer SJ, et al. Charac-terization of 911: a new helper cell line for the titrationand propagation of early region 1-deleted adenoviralvectors. Hum Gene Ther 1996;7:215–22.

27. Malerba M, Daeffler L, Rommelaere J, Iggo RD.Replicating parvoviruses that target colon cancer cells.J Virol 2003;77:6683–91.

28. Amalfitano A, Begy CR, Chamberlain JS. Improvedadenovirus packaging cell lines to support the growth ofreplication-defective gene-delivery vectors. Proc NatlAcad Sci U S A 1996;93:3352–6.

29. Einfeld DA, Schroeder R, Roelvink PW, et al.Reducing the native tropism of adenovirus vectorsrequires removal of both CAR and integrin interactions.J Virol 2001;75:11284–91.

30. Sikorski RS, Hieter P. A system of shuttle vectorsand yeast host strains designed for efficient manip-ulation of DNA in Saccharomyces cerevisiae . Genetics1989;122:19–27.

31. Gagnebin J, Brunori M, Otter M, Juillerat-Jeanneret L,Monnier P, Iggo R. A photosensitising adenovirus forphotodynamic therapy. Gene Ther 1999;6:1742–50.

32. Reich NC, Sarnow P, Duprey E, Levine AJ. Monoclonalantibodies which recognize native and denatured formsof the adenovirus DNA-binding protein. Virology 1983;128:480–4.

33. Dalgaard P. Introductory statistics with R. New York:Springer-Verlag; 2002.

34. Harlow E, Lane D. Antibodies: a laboratory manual.Cold Spring Harbor (New York): Cold Spring HarborLaboratory Press; 1988.

35. Rosin-Arbesfeld R, Cliffe A, Brabletz T, Bienz M.Nuclear export of the APC tumour suppressor controlsh-catenin function in transcription. EMBO J 2003;22:1101–13.

36. Reynolds P, Dmitriev I, Curiel D. Insertion of an RGDmotif into the HI loop of adenovirus fiber protein altersthe distribution of transgene expression of the system-ically administered vector. Gene Ther 1999;6:1336–9.

37. Tao N, Gao GP, Parr M, et al. Sequestration ofadenoviral vector by Kupffer cells leads to a nonlineardose response of transduction in liver. Mol Ther 2001;3:28–35.

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

39. Chen Y, Yu DC, Charlton D, Henderson DR. Pre-existent adenovirus antibody inhibits systemic toxicityand antitumor activity of CN706 in the nude mouseLNCaP xenograft model: implications and proposals forhuman therapy. Hum Gene Ther 2000;11:1553–67.

40. Maretto S, Cordenonsi M, Dupont S, et al. MappingWnt/h-catenin signaling during mouse developmentand in colorectal tumors. Proc Natl Acad Sci U S A 2003;100:3299–304.

41. Gat U, DasGupta R, Degenstein L, Fuchs E. De novohair follicle morphogenesis and hair tumors in miceexpressing a truncated h-catenin in skin. Cell 1998;95:605–14.

42. van de Wetering M, Sancho E, Verweij C, et al. Theh-catenin/TCF-4 complex imposes a crypt progenitorphenotype on colorectal cancer cells. Cell 2002;111:241–50.

43. Liu Q, Zaiss AK, Colarusso P, et al. The role ofcapsid-endothelial interactions in the innate immuneresponse to adenovirus vectors. Hum Gene Ther 2003;14:627–43.

44. Schiedner G, Bloch W, Hertel S, et al. A hemody-namic response to intravenous adenovirus vectorparticles is caused by systemic Kupffer cell-mediatedactivation of endothelial cells. Hum Gene Ther 2003;14:1631–41.

45. Feigenblum D, Schneider RJ. Cap-binding protein(eukaryotic initiation factor 4E) and 4E-inactivatingprotein BP-1 independently regulate cap-dependenttranslation. Mol Cell Biol 1996;16:5450–7.

46. Cuesta R, Xi Q, Schneider RJ. Structural basis forcompetitive inhibition of eIF4G-Mnk1 interaction bythe adenovirus 100-kilodalton protein. J Virol 2004;78:7707–16.

47. Xi Q, Cuesta R, Schneider RJ. Tethering of eIF4G toadenoviral mRNAs by viral 100 k protein drivesribosome shunting. Genes Dev 2004;18:1997–2009.

48. Nemunaitis J, Cunningham C, Buchanan A, et al.Intravenous infusion of a replication-selective adenovi-rus (ONYX-015) in cancer patients: safety, feasibility andbiological activity. Gene Ther 2001;8:746–59.

49. Yang Y, Su Q, Grewal IS, Schilz R, Flavell RA, WilsonJM. Transient subversion of CD40 ligand functiondiminishes immune responses to adenovirus vectors inmouse liver and lung tissues. J Virol 1996;70:6370–7.

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

51. Jain RK. Physiological barriers to delivery of mono-clonal antibodies and other macromolecules in tumors.Cancer Res 1990;50:814–9s.

52. Willett CG, Boucher Y, di Tomaso E, et al. Directevidence that the VEGF-specific antibody bevacizumabhas antivascular effects in human rectal cancer. NatMed 2004;10:145–7.

53. Winkler F, Kozin SV, Tong RT, et al. Kinetics ofvascular normalization by VEGFR2 blockade governsbrain tumor response to radiation; role of oxygenation,angiopoietin-1, and matrix metalloproteinases. CancerCell 2004;6:553–63.

54. Hobbs SK, Monsky WL, Yuan F, et al. Regulation oftransport pathways in tumor vessels: role of tumor typeand microenvironment. Proc Natl Acad Sci U S A 1998;95:4607–12.

55. Ferrara N. Role of vascular endothelial growth factorin physiologic and pathologic angiogenesis: therapeuticimplications. Semin Oncol 2002;29:10–4.

Cancer Research

Cancer Res 2005; 65: (15). August 1, 2005 6890 www.aacrjournals.org

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from

Page 10: RAD001 (Everolimus) Improves the Efficacy of Replicating Adenoviruses … · adenoviruses that replicate selectively in cells with constitutive activation of the Wnt signaling pathway

2005;65:6882-6890. Cancer Res   Krisztian Homicsko, Alexander Lukashev and Richard D. Iggo  Adenoviruses that Target Colon CancerRAD001 (Everolimus) Improves the Efficacy of Replicating

  Updated version

  http://cancerres.aacrjournals.org/content/65/15/6882

Access the most recent version of this article at:

   

   

  Cited articles

  http://cancerres.aacrjournals.org/content/65/15/6882.full#ref-list-1

This article cites 53 articles, 15 of which you can access for free at:

  Citing articles

  http://cancerres.aacrjournals.org/content/65/15/6882.full#related-urls

This article has been cited by 8 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/65/15/6882To request permission to re-use all or part of this article, use this link

Research. on November 26, 2020. © 2005 American Association for Cancercancerres.aacrjournals.org Downloaded from