reovirus: a targeted therapeutic progress and potential · dsrna (5). however, considering the fact...

13
Review Reovirus: A Targeted TherapeuticProgress And Potential Radhashree Maitra 1 , Mohammad H. Ghalib 1 , and Sanjay Goel 1,2 Abstract Medical therapy of patients with malignancy requires a paradigm shift through development of new drugs with a good safety record and novel mechanisms of activity. While there is no dearth of such molecules, one particular agent, "reovirus" is promising by its ability to target cancer cells with aberrant signaling pathways. This double- stranded RNA virus has been therapeutically formulated and has rapidly progressed from preclinical validation of anticancer activity to a phase III registration study in platinum refractory metastatic squamous cell carcinoma of the head and neck. During this process, reovirus has shown safety both as a single agent when administered intratumorally and intravenously, as well as in combination therapy, with multiple chemotherapeutics such as gemcitabine, carboplatin/paclitaxel, and docetaxel; and similarly with radiation. The scientic rationale for its development as an anticancer agent stems from the fact that it preferentially replicates in and induces lyses of cells with an activated Kras pathway. As documented in many previous studies, the initial observation of greater tropism in Kras-compromised situation might certainly not be the sole and possibly not even the predominant reason for enhanced virulence. All the same, scientists have emphasized on Kras optimistically due to its high prevalence in various types of cancers. Incidence of Kras mutation has been found to be highest in pancreatic cancer (85%90%) followed by colorectal (3545%) and lung (2530%). Reovirus, in fact has the potential not only as a therapy but also as a tool to unravel the aberrant cellular pathway leading to carcinogenicity. Mol Cancer Res; 10(12); 151425. Ó2012 AACR. Introduction Exciting research in the past decade has revealed unique viral characteristics not registered in any other microorgan- ism. Viruses harbor distinct strategies to overcome the sophisticated defense mechanisms of the infected host (1). The intricate ability to quickly gain control over the host cellular mechanism has potentially made the virus a double- edged sword. While several viruses have been identied as cancer-causing agents, many have also been identied as therapeutically viable oncolytic elements. One important member of therapeutically identied viruses is Respiratory Enteric Orphan virus commonly known as REOvirus (1). It is naturally oncolytic with inherent propensity to replicate in cells with dysfunctional cell signaling cascade including ras- activation. Preferential oncolytic properties of reovirus can be effectively exploited as clinical therapy due to general mildness of infection often requiring no special medical intervention. The search for oncolytic viruses roots from the fact that transient cancer remission can occur following viral infec- tions (2). Supportive preclinical evidence of the novel mechanisms of anticancer activity of reovirus provided the rationale for therapeutic advancement to clinical trials. Unique mode of tumor destruction encouraged oncolytic viral therapy as potential augmentation of chemotherapy and radiation thus making it a cutting edge clinical approach (3, 4). Many of the oncolytic viruses currently in clinical testing are attenuated derivatives of prevalent human pathogens. Typically, in recent years, they have been genetically engineered to further reduce their pathogenic- ity and increase their oncolytic potency and enhance specicity for cancer tissue. Virus with a double-stranded DNA genome is the most suitable candidate for such manipulations with greater genome stability and lesser chance of hazardous mutations. Adenoviruses and herpes simplex virus are the most suitable and thus been exten- sively engineered. Reverse genetic manipulation of RNA virus is still a scientic challenge even with the availability of the present day cutting edge genetic technology. In a similar vein, reovirus is not amenable to genetic engineer- ing, especially due to its segmented structure of the dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to be tumor cytotoxic, even in the presence of neutralizing antibodies, the lack of engineerability may not be a constraint to its further therapeutic development. Authors' Afliation: 1 Monteore Medical Center, 2 Albert Einstein College of Medicine, Bronx, New York R. Maitra and M.H. Ghalib contributed equally to this work. Corresponding Author: Sanjay Goel, Department of Medical Oncology, Monteore Medical Center, Albert Einstein College of Medicine, 1825 Eastchester Road, Bronx, NY 10461. Phone: 718-904-2488; Fax: 718- 904-2830; E-mail: sgoel@monteore.org doi: 10.1158/1541-7786.MCR-12-0157 Ó2012 American Association for Cancer Research. Molecular Cancer Research Mol Cancer Res; 10(12) December 2012 1514 on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Upload: others

Post on 01-Jan-2021

2 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

Review

Reovirus: A Targeted Therapeutic—Progress And Potential

Radhashree Maitra1, Mohammad H. Ghalib1, and Sanjay Goel1,2

AbstractMedical therapy of patients withmalignancy requires a paradigm shift through development of new drugs with a

good safety record and novel mechanisms of activity. While there is no dearth of such molecules, one particularagent, "reovirus" is promising by its ability to target cancer cells with aberrant signaling pathways. This double-stranded RNA virus has been therapeutically formulated and has rapidly progressed from preclinical validation ofanticancer activity to a phase III registration study in platinum refractory metastatic squamous cell carcinoma of thehead and neck. During this process, reovirus has shown safety both as a single agent when administeredintratumorally and intravenously, as well as in combination therapy, with multiple chemotherapeutics such asgemcitabine, carboplatin/paclitaxel, and docetaxel; and similarly with radiation. The scientific rationale for itsdevelopment as an anticancer agent stems from the fact that it preferentially replicates in and induces lyses of cellswith an activated Kras pathway. As documented in many previous studies, the initial observation of greater tropismin Kras-compromised situation might certainly not be the sole and possibly not even the predominant reason forenhanced virulence. All the same, scientists have emphasized on Kras optimistically due to its high prevalence invarious types of cancers. Incidence of Krasmutation has been found to be highest in pancreatic cancer (85%–90%)followed by colorectal (35–45%) and lung (25–30%). Reovirus, in fact has the potential not only as a therapy butalso as a tool to unravel the aberrant cellular pathway leading to carcinogenicity.Mol Cancer Res; 10(12); 1514–25.�2012 AACR.

IntroductionExciting research in the past decade has revealed unique

viral characteristics not registered in any other microorgan-ism. Viruses harbor distinct strategies to overcome thesophisticated defense mechanisms of the infected host (1).The intricate ability to quickly gain control over the hostcellular mechanism has potentially made the virus a double-edged sword. While several viruses have been identified ascancer-causing agents, many have also been identified astherapeutically viable oncolytic elements. One importantmember of therapeutically identified viruses is RespiratoryEnteric Orphan virus commonly known as REOvirus (1). Itis naturally oncolytic with inherent propensity to replicate incells with dysfunctional cell signaling cascade including ras-activation. Preferential oncolytic properties of reovirus canbe effectively exploited as clinical therapy due to generalmildness of infection often requiring no special medicalintervention.

The search for oncolytic viruses roots from the fact thattransient cancer remission can occur following viral infec-tions (2). Supportive preclinical evidence of the novelmechanisms of anticancer activity of reovirus provided therationale for therapeutic advancement to clinical trials.Unique mode of tumor destruction encouraged oncolyticviral therapy as potential augmentation of chemotherapyand radiation thus making it a cutting edge clinical approach(3, 4).Many of the oncolytic viruses currently in clinical

testing are attenuated derivatives of prevalent humanpathogens. Typically, in recent years, they have beengenetically engineered to further reduce their pathogenic-ity and increase their oncolytic potency and enhancespecificity for cancer tissue. Virus with a double-strandedDNA genome is the most suitable candidate for suchmanipulations with greater genome stability and lesserchance of hazardous mutations. Adenoviruses and herpessimplex virus are the most suitable and thus been exten-sively engineered. Reverse genetic manipulation of RNAvirus is still a scientific challenge even with the availabilityof the present day cutting edge genetic technology. In asimilar vein, reovirus is not amenable to genetic engineer-ing, especially due to its segmented structure of thedsRNA (5). However, considering the fact that thisnaturally oncolytic, and pathologically self resolving virusseems to be tumor cytotoxic, even in the presence ofneutralizing antibodies, the lack of engineerability maynot be a constraint to its further therapeutic development.

Authors' Affiliation: 1Montefiore Medical Center, 2Albert Einstein Collegeof Medicine, Bronx, New York

R. Maitra and M.H. Ghalib contributed equally to this work.

Corresponding Author: Sanjay Goel, Department of Medical Oncology,Montefiore Medical Center, Albert Einstein College of Medicine, 1825Eastchester Road, Bronx, NY 10461. Phone: 718-904-2488; Fax: 718-904-2830; E-mail: [email protected]

doi: 10.1158/1541-7786.MCR-12-0157

�2012 American Association for Cancer Research.

MolecularCancer

Research

Mol Cancer Res; 10(12) December 20121514

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 2: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

Preclinical Studies with ReovirusReovirus as a single agentIn vitro data with reovirus. Several preclinical studies

documented oncolytic characteristics of reovirus (6, 7).Initial studies with NIH-3T3 cells revealed that resistanceto reovirus infectivity can be overcome by transformationwith activated Sos/Ras oncogenes (8, 9). The findings indi-cated that usurpation of Ras signaling pathway constitutesthe basis of viral oncolysis (8). Several in vitro studies withhuman cancer cell lines documented the evident role ofreovirus in cellular cytopathy (4, 8). A systematic analysis ofthe propensity of reovirus infectivity towards 24 establishedglioma cell lines was conducted. Dramatic and widespreadcell killing after exposure to live (but not dead) reovirusoccurred in 20 (83%) cell lines. After 48 hours of infection,widespread cell death was found in U87, U251N, and A172cell lines, and almost complete cell death was seen after72 hours. In contrast, cells receiving either dead or no virusremained healthy. Furthermore, to ensure that cell lysis wasdue to viral replication, cells were reacted with rabbit anti-reovirus antibody, followed by FITC-conjugated goat anti-rabbit IgG when susceptible lines depicted the expressionof the viral antigens. Replication of reovirus in susceptiblelines was further confirmed by [35S]methionine metaboliclabeling (4).Short-term primary culture from surgically excised human

glioma have shown100% sensitivity to reoviral oncolysis (4).Studies conducted to test the ability of reovirus to infect andkill primary cultures of brain tumors freshly excised from 9patients with glioma intriguingly depicted complete growtharrest and cell death (100%), although similar level ofreovirus-mediated cellular cytopathy was not documentedwith 7 primary meningioma cultures (4). The ability ofreovirus to lyse all primary glioma cell cultures derived fromsurgical specimen suggested that a substantial proportion ofgliomas may respond to reovirus treatment. It can be arguedthat the number of specimens being relatively small, theobservations might not reflect the comprehensive tumorproperties of gliomas. The oncolytic spectrum of reovirus hasalso been studied in 6 breast cancer cell lines where a highsusceptibility was documented (10). An infectivity of 10MOI (multiplicity of infection) was used in the study.Furthermore, the controlHs578Bst normalmammary glandepithelial cell line used in the study did not show anycytopathic effect (CPE) to the virus confirming the fact thatreovirus preferentially targets the transformed cells sparingthe normal ones (9). Contemporary studies with melanomacell lines and primary cultures from fresh resected tumorsrevealed similar results. The transformed cells allowed viralreplication followed by caspase-dependent cytotoxicity (11).Normal melanocytes conversely resisted viral replication.Notably, it was also reported that all of the screened breastcancer and melanoma cell lines had activated Kras.The basis of the ability of reovirus to target and kill tumor

cells but not infect nonproliferating normal cells lies in itsability to usurp the highly activated signaling pathway foundin tumor cells (4). This ability is most clearly established forRas or elements in its downstream pathways.Ras activation is

very common in malignant gliomas, colorectal (CRC) can-cers, as well as pancreatic malignancies.Natural affinity of reovirus to kill and lyse cancer cells and

the prevalence of Kras mutation in many of the studiedmodels apparently instigated the researchers to attempt todraw a correlation between Kras mutation and viral onco-lysis. It is to be noted that the fact is yet to be substantiatedwith scientific evidence. A somewhat similar situation wasfaced by the scientific community while developing thetherapeutically viable adenovirus Onyx-01. The antitumoractivity of the virus was initially proposed to be solelydependent on the status of p53 but deeper introspectionrevealed that lytic activity was observed in both p53 negativeand WT conditions and the complex molecular mechanismof virus mediated oncolysis is yet to be clearly defined (12).Further studies to elucidate cellular events favoring reovi-

rus-mediated apoptosis confirmed that colon cancer celllines, HEK293 and HCT116 displayed elevated b-cateninexpression to promote reovirus-mediated oncolysis by down-regulation of NF-kB (3). Independent studies reported thatreovirus activates human dendritic cells to promote innateantitumor immunity (13). The exact role of different immuneeffector cells in oncolytic virus–mediated tumor regression hasnot yet been clearly defined. It is plausible that dendriticcells (DC) are likely to play a coordinating role in virus-mediated immune response as key antigen presenting cells(APC) that recognize the viral infection and regulate bothinnate and adaptive immunity (11). The fact that reovirus hasbeen found to be effective in tumor cytopathy despite increas-ing neutralizing anti reovirus antibody (NARA) in the serumlogically indicates the intricate role of the innate immunesystem in the virus-mediated oncolytic process. The obser-vation that reovirus-activatedDCs enhance the innate naturalkiller (NK) cells and cytotoxicT cells (Tc) by release of solublefactors inducing tumor cell killing via exocytosis clearlysupports the hypothesis. The role of NK cells in tumorregression has been well documented in mouse model bothby direct tumor recognition as well as via DC activation (14).Reovirus-induced DC maturation also stimulated the pro-duction of proinflammatory cytokines interferon (INF)-a,tumor necrosis factor (TNF)-a, interleukin (IL)-12p70, andIL-6. Activation of DCs by reovirus was not dependent onviral replication, whereas cytokine production was inhibitedby the blockade of PKR (protein kinase receptor) and NF-kBsignaling. These observations provide a hint that reovirus-mediated DC activation and the downstream immune sig-naling is multimodal. Although systemic delivery activatesthe adaptive immune system and triggers a robust antibodyresponse, intratumoral (ITu) injection of the virus results insuccessful tumor destruction by activation of the innateimmune effector cells within the tumor microenvironment.Hence, reovirus recognition by DCs may trigger innateeffecter mechanisms to complement the virus's direct cyto-toxicity, potentially enhancing the efficacy of reovirus as atherapeutic agent (13). Intravenous administration of reovirusin conjunction with immunosuppressants has been found tobe therapeutically more effective indicating that the neutral-izing antibodies are not completely blunted (15). All the same,

Development of Reovirus for Cancer Therapy

www.aacrjournals.org Mol Cancer Res; 10(12) December 2012 1515

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 3: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

the fact that antitumor activity is observed even in thepresence of virus-neutralizing antibodies can also indicate aplausible role of the host cellular machinery in camouflagingthe virus and thus preventing its recognition by the specificantibodies. In a very elegantly conducted translationalresearch study, it has been shown that replicating virus isdetectable in cellular compartment of the blood, namely inmononuclear, granulocyte and platelets, and this may be amechanism of protection from NARA (16). Furthermore, inthis same clinical trial, when patients' tumors were harvested,replicating virus was evident in tumor tissue but not fromnormal liver, showing some evidence of selective tropism formalignant cells.PKR plays crucial inhibitory role in efficient viral

replication essential for infective virion production andoncolysis (refs. 7, 17; Fig. 1). Expression of PKR isupregulated in response to INF released by infected cells(7). Binding to viral RNA/initial transcripts results in PKRdimerization, autophosphorylation, and activation (7).The viral S1 segment mRNA has been shown to be apotent activator of PKR (18). Once activated, PKR blocksthe primary and secondary reovirus protein translation.In Ras-transformed cells, PKR is not activated, allowingunabated viral replication and effective assembly of viralproteins for production of infection efficient virions(7, 17). Specific chemical inhibitors of PKR phosphory-lation restore reovirus translation in untransformed cellsproviding evidence for a direct role of PKR in definingresistance to reovirus replication (9). Ras activation issuggested to release the translational blocks in the trans-formed cells. However, the exact molecular mechanism ofcoordination between Ras activation and inhibition ofPKR-mediated viral translational remains elusive (Fig. 1).The recently discovered novel oncogene CUG2 (cancer

upregulatory gene-2), inhibits the expression of PKR andactivatesRas and p38MAPK (19). Studies further confirmedthat inhibition of p38 MAPK or Ras blocks reoviral pro-liferation even in the presence of CUG2 indicating thepossibility of multimodal crosstalk between Ras activationand inhibition of PKR phosphorylation (19). It is pertinenttomention that PKR dimerization and autophosphorylationis critical for effective viral propagation but the definitivecontribution of Kras mutation in the process is still elusive.Aberrant cell signaling cascade generates many anomalousevents within the cell and exactly how it facilitates theinhibition of PKR dimerization is yet to be defined clearly.Investigation of the mechanism of apoptosis in reovirus-

infected HEK293 cell line concluded a significant role ofTRAIL (TNF-related apoptosis-inducing ligand). The apo-ptotic event was successfully inhibited by anti-TRAIL anti-bodies or death receptor (DR)4 and DR5 (20). Similarinvolvement of TRAIL was confirmed in cell lines derivedfrom 2 different human lung (A157/H549) and breastcancers (MDA231/ZR75-1; refs. 20, 21). Reovirus infectionsynergistically sensitizes these cancer cell lines to killing byexogenous TRAIL. The observed sensitization was associ-ated with an increase in the activity of the death receptor-associated initiator caspase-8, and was inhibited by the

peptide IETD-fmk, suggesting that reovirus sensitizescancer cells to TRAIL-induced apoptosis in a caspase-8–dependent manner. Enhanced sensitization was also foundto be associated with increased cleavage of PARP, a substrateof the effector caspases 3 and 7 (22).In vivo experience using animal models of cancer.

Tumor regression studies in animals with reovirus as singleagent showed dramatic results. Pronounced effects oftenwith complete tumor regression was documented in vivo in 2subcutaneous (P ¼ 0.0002 for both U251N and U87) andin 2 intracerebral (P ¼ 0.0004 for U251N and P ¼ 0.0009for U87) human malignant glioma mouse models (4).Immunocompetent C3 mice implanted with Ras-trans-formed C3H10t1/2 fibroblast showed complete regressionof tumor after ITu injection of reovirus (23). Similar strategyof ITu injection resulted in significant tumor regression inSCID mice with subcutaneously implanted v-erbB–trans-formed NIH-3T3 cells (23), and in SCID-NOD mice withsubcutaneously implanted human malignant glioma (4, 8).Subcutaneous tumor allograft studies in immune competentC3H mice showed strong inhibition of tumor growth withintravenous administration of reovirus (15). A 74% cure ratewas observed in comparatively less immunocompromisednude mice harboring human malignant glioma with singleintralesion injection of reovirus. SCID-NOD mice bearingsubcutaneous U251N xenografts (4) or multiple myelomaRPMI8226GFPþ cells introduced via tail vein for tumorestablishment, when treated with single injection of live ordead reovirus showed a striking regression of live virus–treated tumors (24). Immunofluorescence analysis showedthat reovirus replication was restricted to the tumor masswithout spreading to the underlying normal tissue.

Reovirus as combination therapyAssociation with radiotherapy. Combination therapy

involving radiation and reovirus was evaluated both in vitroand in vivo (21). The CRC cell line HCT116 was treatedwith reovirus and radiation individually, and in combina-tion, showed a marked synergy in the combinational subset.In vivomurine studies depicted similar synergy in nude micewith tumor induced by HCT116/SW480 cell implantationas well as in C57BL6 mice by B16 melanoma cells mediatedtumor induction (21).The relative tumor volume (RTV) whenmeasured in nude

mice xenografted with RH30 and SK-ESI human sarcomaand treated with similar doses revealed lowest mean tumorvolume and longest event-free survival in the group receivingthe combination treatment. TUNEL assay conducted withtumor biopsies showed enhanced apoptotic activity in com-bination therapy as compared with single agent (25).Association with chemotherapy. In vitro studies to

determine augmentation of viral therapy with chemotherapywas done using gemcitabine, fluorouracil, cisplatin, anddoxorubicin in HCT116 cells. Reovirus was synergisticwith all 4 drugs across a wide range of concentration (26).The synergistic consequences observed with gemcitabineadvanced further onto in vivo evaluation of xenograftednude mice. A remarkable synergy was confirmed with no

Maitra et al.

Mol Cancer Res; 10(12) December 2012 Molecular Cancer Research1516

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 4: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

residual tumor tissues remaining at the end point of the studyin more than 95% of the mice (27).Another independent study of reovirus in combination

with cisplatin, mytomycin, vinblastine, or gemcitabine inNSCLC cells lines documented synergy in cell lines sensitiveto the chemotherapeutic drugs and antagonism in cell linesthat are drug resistant. The taxanes showed reasonablesynergy when administered in combination even in drug-resistant cell lines (28). Similarly C57BL/6 rodent with B16melanoma–induced tumor when treated by combinationtherapy of reovirus and cisplatin showed a synergistic reduc-

tion in RTV as compared with those receiving monotherapy(29).In context of animal models of human tumors, athymic

mice bearing xenografts of osteosarcoma, Ewings, and syno-vial sarcoma when treated in similar fashion also confirmed asynergistic antitumor activity in comparison with mono-therapy (30). Docetaxel administered in combination in celllines and PC-3 prostate cancer mouse model confirmedsynergy, which diminished with increasing docetaxel con-centration (31). The confinement of viral proteins within thetumor mass was confirmed by several studies including

Reovirus

Kra

s W

T

Kras m

utant

GAP RAS

RAS

GDP

GTP

GEF

GAP

RAS

??

RAS

GDP

GTP

GEF

Phagocytosis

Endosome

Viraluncoating

CUG2

Translation blocked

Translation

Cell lysis

release of infective virions

Protein

Virus assembly

Transcripts

P-PKR

P-elF2α elF2α

PKR

ds-RNA

Figure 1. A schematic illustration of the current hypothesis of the fate of reovirus upon entering the host cell by phagocytosis. The status of the host cellsignaling cascadedetermines the downstreamconsequences.Kras (Kirsten rat sarcoma viral Oncogene) is amembrane-boundmolecular switchwhich bindsto and catalyses the hydrolysis of GTP (guanosine triphosphate) to initiate a cell signal. The hydrolysis is facilitated by GTPase activating protein(GAP) and guanine nucleotide exchange factor (GEF) with subsequent conversion to inactive Kras GDP (guanosine diphosphate) complex. On the left isrepresented the k-raswild-type (WT) environment, which prevents the translation of viral transcripts and abrogates reovirus assembly. The double-strandedRNA (dsRNA)-dependant protein kinase R (PKR) plays a crucial role in the viral translation block by dimerization and autophosphorylation to phospho-PKR(P-PKR), which in turn promotes the phosphorylation of translation initiation factor 2 subunit alpha (eIF2a) to P-eIF2a. The dashed arrow depicts thetranslational inhibition by P-eIF2a in Kras WT cells. In Kras-compromised situation (shown on right), the autophosphorylation of P-PKR is inhibited withunabated viral translation and subsequent virion assembly and cell lysis. The factors contributing to the crosstalk between defective Kras and compromisedPKR phosphorylation are unknown and thus represented by (?). Cancer upregulatory gene 2 (CUG2) frequently elevated in variety of tumor tissues hasbeen recently characterized to promote reoviral proliferation by activation of Kras and inhibition of PKR phosphorylation. CUG2 has been proposed as abiomarker to identify the subset of cancers that might benefit from reoviral therapy.

Development of Reovirus for Cancer Therapy

www.aacrjournals.org Mol Cancer Res; 10(12) December 2012 1517

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 5: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

immunohistochemical evaluation of U87 xenograftedrodent (8). The broad oncolytic spectrum of reovirus assingle agent and in combination along with remarkablesuccess of the animal study platform provided the scientificconfidence to proceed onto clinical trials. Although muchpreclinical work has been carried out establishing thatreovirus could serve as a cancer therapy, the mechanismsgoverning the permissiveness of transformed cells to reovirusinfection remain to be fully characterized. Furthermore, asreovirus replicates in cancers of very diverse origin, it is likelythat the virus exploits cellular signals that occur often intransformation and tumorigenesis. A deeper understandingof the molecules involved in antiviral defense and thepatterns they recognize will allow harnessing them for bettertherapeutic strategies. Thus, delineating this usurpationshould in turn shed light on signaling that is commonamong various cancers, and may reveal novel therapeutictargets.Although the first hint of reovirus susceptibility (32) was

noted in 1977, it was not until 1990s that any scientific cluewas obtained regarding the preferential replication of reo-virus in transformed cells. Since then, systematic researchhas provided some clues on the molecular synergy of effi-cient reovirus oncolysis in ras-compromised cancer cells.Although the preliminary findingmight not be substantial indefining the viral tropism, it has nevertheless logicallypermitted clinicians to attempt the use of reovirus as treat-ment towards ras-mutated carcinogenicity especially in sit-uation where no other well established treatment regime isavailable.Incidentally, reovirus is intrinsically oncolytic without

need for any genetic manipulation. It is a naturally occurringvirus that possesses the right level of potency that renders itrelatively harmless to normal cells while having strong lyticeffect on ras-compromised cancer cells. Investigations on themechanisms of selective viral oncolysis in ras-mutated tumorcells will help unravel the complex cellular pathways involvedin cellular transformation which in turn will not only help inidentifying novel targets for cancer therapy but also shedlight on which cancer backgrounds are compatible with viraloncolysis strategies.

Clinical Experience with ReovirusReovirus has been therapeutically tested in 325 (as

reported) patients administered ITu or intravenously eitheras a monotherapy or in combination with radiotherapy orchemotherapy. The first human trial with reovirus began inJuly 2001, and since then a total of 27 clinical trials havebeen completed/initiated/planned as phase 1, 2 and 3.Thirteen trials have been completed, whereas 12 are ongoingand 2 have been announced. Of these 27 trials, 6 will besponsored by the National Cancer institute. Of the 16 trialswith reported clinical data, clinically formulated reovirus wasinjected ITu in 6 trials enrolling 90 patients, whereas theremaining 10 clinical trials involved the intravenous routeenrolling 235 patients (Table 1). The current dosing regi-men is intravenous of 3� 1010 TCID50 on days 1 to 5 over60 minutes repeated every 3 or 4 weeks in both mono- and

combination therapy. As with the preclinical observations,the clinical experience with reovirus has been interesting,evolving, and has shown promise. Summaries of the resultsare provided in Table 1.

Reovirus as intratumoral injectionsThe first human phase 1 trial was conducted in 18 patients

with metastatic or recurrent solid tumors with easy access tothe tumor to allow for direct ITu injection, and measure-ment by direct observation or palpation. The treatment waswell tolerated without any observation of dose-limitingtoxicity (DLT) and/or maximum tolerated dose (MTD;ref. 33). The second trial was based on the preclinicalexperience of reovirus on prostate-derived cell lines (34).Six patients with prostate cancer (stage T2/T3) received asingle transrectal ultrasonography (TRUS)-guided reovirusinjection followed by radical prostatectomy after 3 weeks.Histologic analysis after prostatectomy of the injected lesionsand other synchronous lesions showed CD8 T-cell infiltra-tion and evidence of caspase-3 activity within the reovirus-injected areas. The third clinical trial was based on thepreclinical data showing reovirus activity in malignant gli-omas (4). While oncolytic viruses have been used in clinicaltrials in malignant gliomas (35–38), this study was the firstto use reovirus. A total of 12 patients (10 glioblastomamultiforme, 1 anaplastic astrocytoma, and 1 anaplasticoligoastrocytoma) were treated at 3 dose-escalating levels(ongoing trial). Antibody studies showed a seroconversionrate of 83% consistent with other clinical studies. Subse-quently, a combined phase 1/2 study (currently ongoing)was started in recurrent malignant gliomas with a singleITu infusion (using transcranial catheters) of reovirusdirectly into the intracerebral tumor over 72 hours.Accrual to the phase I portion has been completed, butthe data are pending.In another 2-stage phase I trial, dose-escalating reovirus

was given ITu along with palliative radiotherapy to 23patients with advanced cancers (39). There was no evidenceof exacerbation of skin toxicity induced by radiation. Therewas marked efficacy with 7 of 14 evaluable patients havingexperienced a partial response (PR). In another phase 2,multicenter, clinical trial, ITu reovirus was evaluated whencombined with low-dose radiotherapy (20 Gy) in patientswith advanced cancer (40). This study showed safety andefficacy of the combination with 4 patients with PR and 2with minimal response.

Reovirus as systemic administrationThere have been 2 single agent, phase 1 trials of reovirus

as a systemic intravenous infusion. One clinical trial wasconducted at our center, MontefioreMedical Center, AlbertEinstein College of Medicine (Bronx, NY; ref. 41). This wasa single center dose-escalation trial and enrolled 18 patientswith refractory solid tumors. Overall, toxicities were minor,with only 2 patients experiencing grade 2 events, withoutan observation of a single DLT. One patient with breastcancer who received 7 cycles developed grade 2 fever andgrade 1 chills that progressively worsened with repeated

Maitra et al.

Mol Cancer Res; 10(12) December 2012 Molecular Cancer Research1518

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 6: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

Tab

le1.

Sum

maryof

clinical

trialswith

reporteddata.

Thistable

summarizes

thecu

rren

tdatafrom

completedan

don

goingclinical

trialswith

Reo

virus

Study#

Indication

Other

therap

yNum

ber

of

patients

Pha

se

Lowes

tdose

(TCID

50)

Highe

stdose

(TCID

50)

Grade3/4

toxicities

Effica

cy

Viral

detec

tion

serum/C

SF/

stool/urine/

fece

sAntibody

resp

ons

eReferen

ceCommen

ts

Intratum

oral

1Solid

tumors

181

107(PFU

)10

10(PFU

)Non

e1CR,1

PR,

8SD

Serum

Yes

33

2Prostate

6Pilo

t10

7(PFU

)Non

e1PR,4

SD

Urin

eYes

343

Maligna

ntgliomas

121

1�

107

1�

109

Tran

sien

tGGT

elev

ation

1SD

Saliva,

Fece

sYes

6Solid

tumors

XRT

231

1�

108

1�

1010

Non

e7PR,7

SD

Non

eYes

3920

–36

Gy

7Maligna

ntgliomas

151/2

1�

108

1�

1010

NR

NR

NR

NR

8Solid

tumors

XRT-20

Gy

162

1�

1010

Non

e4PR,2

MR,

7SD

NR

Yes

40

Intrav

enou

s4

Solid

tumors

181

1�

108

3�

1010

Non

e1PR,7

SD

Urin

e,Serum

,Saliva,

Fece

sYes

41

5Solid

tumors

331

1�

108

3�

1010

Flu-likesy

ndrome

3MR,7

SD

Urin

e,Serum

,Saliva,

Fece

sYes

44

CPK-M

B/TropI

elev

ation

Lympho

pen

iaNeu

trop

enia

9Solid

tumors

Gem

citabine

1000

mg/m2

121

1�

109

3�

1010

Flu-like

synd

rome

48

Neu

trop

enia

AST/ALT

/GGT

elev

ation

Trop

Ieleva

ton/

STch

ange

s10

Solid

tumors

Doc

etax

el75

mg/m2

241

1�

109

3�

1010

Febrile

neutropen

ia4PR,3

MR,

7SD

Urin

e,Serum

,Saliva,

Fece

sYes

54

Thromboc

ytop

enia

GIs

ymptoms

Hyp

okalem

iaHyp

oten

sion

NR

NR

11Solid

tumors

Carbop

latin

AUC

531

1/2

3�

109

3�

1010

Mye

losu

ppress

ion

8PR,6

SD

56Effica

cydata

amon

g19

Pac

litax

el17

5mg/m

2

Hyp

oten

sion

patientswith

Hea

dan

dNec

kCan

cer

(Con

tinue

don

thefollo

wingpag

e)

Development of Reovirus for Cancer Therapy

www.aacrjournals.org Mol Cancer Res; 10(12) December 2012 1519

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 7: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

Tab

le1.

Sum

maryof

clinical

trialswith

reporteddata.

Thistable

summarizes

thecu

rren

tdatafrom

completedan

don

goingclinical

trials

with

Reo

virus

(Con

t'd)

Study#

Indication

Other

therap

yNum

ber

of

patients

Pha

se

Lowes

tdose

(TCID

50)

Highe

stdose

(TCID

50)

Grade3/4

toxicities

Effica

cy

Viral

detec

tion

serum/C

SF/

stool/urine/

fece

sAntibody

resp

ons

eReferen

ceCommen

ts

13Colorec

tal

canc

erwith

liver

metas

tase

s

102

1�

1010

Yes

16Viru

sse

lectively

detec

ted

inca

ncerou

sliver

tissu

e14

Sarco

mas

532

3�

1010

Neu

trop

enia

19SD

NR

NR

Optic

neuritis

15Hea

dan

dne

ckca

ncer

Carbop

latin

AUC

514

23�

1010

Neu

trop

enia

4PR,2SD

NR

NR

57

Pac

litax

el17

5mg/m

2

Hyp

okalem

iaFa

tigue

Ane

mia

Nau

sea

Hyp

oten

sion

16NSCLC

Carbop

latin

AUC

622

23�

1010

Mye

losu

p-

pression

6PR,13

SD

NR

NR

58

Pac

litax

el20

0mg/m

2

Hyp

oten

sion

Fatig

ueGIs

ymptoms

17Pan

crea

ticca

ncer

Gem

citabine

800mg/m

2

182

3�

1010

Neu

trop

enia

1un

confi

rmed

PR

NR

NR

52

7SD

NOTE

:The

stud

y#prece

dedby"R

EO"indicates

thenu

mber

assign

edbythestud

ysp

onso

r,na

melyOnc

olytics,to

enab

lean

easy

search

forthe

read

er.W

hileso

metrialsaremature

andfullreports

have

bee

npub

lishe

d,o

thersareon

goingwith

prelim

inaryda

taprese

nted

atinternationa

land

natio

nalm

eetin

gsor

updated

ontheco

mpan

y'sweb

site.T

hetable

provides

aquick

overview

with

focu

son

thestud

ydes

ign,patient

accrua

lnum

bers,do

sing

,immun

olog

y,toxicitie

s,an

deffica

cy,and

commen

tsab

outthe

unique

ness

oftheco

nduc

tor

theresu

ltsof

thestud

y.Abbreviations

:AUC:a

reaun

der

theco

ncen

trationtim

ecu

rve;

XRT:

radiatio

n;MR:m

inim

alresp

onse

;NR:n

otreported;P

FU:p

artic

leform

ingun

its.

Maitra et al.

Mol Cancer Res; 10(12) December 2012 Molecular Cancer Research1520

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 8: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

administration. She had a PR and her tumor had a mutationin ras codon 12 (42, 43). In the second, 33 patients enrolled,of which 10 patients showed stable disease (SD; ref. 44).Overall, 28 patients showed an increase in NARA titers to amaximum at 4 weeks, which remained constant duringsubsequent cycles (45). Despite the NARA response, whichcould have blunted the viral delivery to tumor site (via rapidneutralization), viable virus was shown in post treatmentbiopsies after 2 cycles (45).More recently, in a phase 2 study that enrolled patients

with advanced CRCwith liver metastasis, reovirus was givenintravenously for 5 consecutive weeks before the plannedliver metastesectomy. Among the 10 patients treated so far,there is evidence that reovirus selectively targets tumor cellsversus normal liver cells (60% of patients have shown noevidence of reovirus in their normal liver cells). In 2 patients,only necrotic tissue was seen, whereas reovirus was detectedin the immune cells of the tumor of 1 patient. This newexciting information shows that reovirus can be deliveredspecifically and selectively as a monotherapy while sparingthe normal liver cells (ongoing trial). The first completedphase II trial of single agent reovirus targeted 53 patients withsoft tissue and bone sarcomas withmetastasis to lung. A totalof 19 patients showed SD. One patient with synovialsarcoma had SD for more than 80 weeks.There is strong preclinical rationale to combine intrave-

nous infusion of reovirus with cytotoxic chemotherapy likegemcitabine, carboplatin, or docetaxel (15, 27, 28, 46, 47).The phase 2 dose of reovirus was based on two-phase 1monotherapy trials (41, 44). It was postulated that chemo-therapy might blunt the immune-mediated viral clearancewithout significantly increasing the toxicity profile. In thefirst combination phase I study, intravenous reovirus (on day1–5 of each cycle with the starting dose at 3� 109 TCID50)was administeredwith gemcitabine at 1,000mg/m2 on day 1and day 8 (21-day cycle) in dose-escalating cohorts (48).However, with the observation of 2 DLTs [grade 3 ALT(alanine aminotransferase) rise and grade Troponin I] thedose of reovirus was amended to a 1-day treatment. Onepatient with nasopharyngeal carcinoma [(NPC) which iscaused by an Epstein Barr virus (EBV) infection] showed aPR, possibly due to the expression of EGFR (EBV-inducedexpression). The combination showed clinical activitydespite the potential NARA response, and ongoing workis unraveling the intricate details of mechanisms involved inreovirus-medicated oncolysis and resistance (49, 50). On thebasis of this clinical (48) and prior preclinical findings (51), aphase 2 clinical trial in patients with metastatic pancreaticcancer is ongoing, and preliminary data suggests a clinicalbenefit rate of 58%with prolonged SD (52). Preclinical datasuggestive of hepatic toxicity associated with reovirus (53)and the observation of the ALT elevation when combinedwith gemcitabine (48) should be factored in dealing withsituations where patients have compromised hepatic func-tion or are taking potentially hepatotoxic medications, suchas acetaminophen.Therapeutically formulated reovirus has been combined

with docetaxel in 24 patients with advanced cancer (54). Of

all patients treated, 46% experienced grade 3 or greaterneutropenia, consistent with that observed with docetaxelmonotherapy (65%) at the same dose and schedule. MTDwas not technically reached as only one DLT, of grade 4neutropenia, was encountered. Four PR were observed inbreast, stomach, gastroesophageal, and ocular melanomaalong with 3 minor responses in mesothelioma, prostatecancer, and squamous cell cancer of the head and neck. Only2 patients showed evidence of viral shedding. Pharmacoki-netic studies revealed no change in docetaxel clearance by theaddition of reovirus. Docetaxel showed no effect on NARA,a finding that was inconsistent with preclinical data (55).In a separate phase I trial, reovirus was added to the

combination of carboplatin and paclitaxel (56). With pre-liminary signs of clinical activity, the phase 2 expansioncohort was enriched with patients with squamous cellcarcinomas of head and neck (HNSCC), and remarkably,8 of 19 (42%) evaluable patients showed PR. The toxicitieswere consistent with earlier results. This study showed thesafety and efficacy of this combination, leading to a targetedphase II study in patients with HNSCC with a RR of 31%(57), and finally to a randomized phase III study of carbo-platin and paclitaxel with orwithout reovirus in patientswithplatinum refractory HNSCC (ongoing trial). The samecombination has also been tested in patients with advancednon–small cell lung cancer (58). As reported at the 14thWorld Lung Cancer Congress, the combination was welltolerated and of the 23 patients entered, 6 patients had a PRand 13 had SD.

Neutralizing anti-reovirus antibodiesAn important and critical issue when considering the

clinical use of reovirus is the presence of preexisting andthe development of a rapid massive rise of titer of "ontherapy" NARA. This phenomenon of NARA developmenthas been consistently observed across the single agent and inthe chemotherapy combination trials. It is clearly evidentthat despite the presence of preexisting NARA, there is adetectable 100-fold increase in the viral titer, which has beensuggested to be both desirable and unwarranted. On onehand, it allows for limiting the toxic effects of the virus andprotects patients from the unwanted side effects of the virusinfection, whereas, on the other, it may compromise itsbeneficial potent anticancer effects. Prior preclinical in vivostudies in murine models using C3H mice have shown thatblunting the immune response using cyclosporine A or antiCD4/CD8 antibodies leads to improved efficacy (15).Similarly, cyclophosphamide, when used as a modulatingagent, was effective in ensuring tumor seeding of the intra-venously delivered reovirus in tumors, an entity which waspreviously not attainable (59). By modifying the dose of thecyclophosphamide, the authors also showed that it modu-lates, but does not ablate, the NARA response.The concomitant delivery of reovirus with chemotherapy

has been purported to be of benefit, partly by the attenua-tion of the NARA response. However, in combinationchemotherapy trials, specifically with docetaxel, and carbo-platin–paclitaxel, there was a significant rise in NARA titer

Development of Reovirus for Cancer Therapy

www.aacrjournals.org Mol Cancer Res; 10(12) December 2012 1521

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 9: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

and no discernable increase in toxicity (46, 56). Anotherinteresting phenomenon observed in these trials was that incomparison with the single-agent studies, the rate of rise ofthe NARA titer was lower, thereby leading to a delay in theachievement of the peak titer. Slower development ofNARA may have beneficial effects by enhancing tumorseeding of the virus leading to greater efficacy withoutcompromising safety. Interestingly, the only agent that hadsome suggestion of the ability to blunt the NA response wasgemcitabine (48). This is not surprising as gemcitabine hasbeen shown in murine models to specifically affect thegeneration of antibodies by B cells (49) and to suppressmyeloid suppressor cells (60). Furthermore, independent ofthe attenuation of theNARA response, gemcitabine has beenreported to tip cellular immunity favoring reovirus-initiatedantitumor immune response (51, 60). The attenuatedNARA response allowed for greater virus replication leadingto greater toxicity of the combination as compared with thesingle-agent profile, an important but not entirely unex-pected clinical observation. Therefore, careful monitoring ofimmune function should remain an important considerationin future combination trials.Moreover, it has been suggestedthat because of the interference of the adaptive immunity ofthe host, the most effective systemic delivery of reovirus willbe achieved through rapid, repeated high doses of viruswithin the first week of treatment before theNARA responsehas been boosted (45). The current dosing schema of 5continuous daily intravenous injections of the virus every 3to 4 weeks, therefore, has some clinical and scientificrationale.

Summary, Conclusions, and the FutureIn summary, reovirus has shown promise with far reaching

implications for future drug development. While initiallydeveloped as an anticancer agent based on the scientificpremise that viral replication is supported in ras-drivencancer cells; the paradox lies in that the clinical developmenthas not been driven by this fact. Moreover, clinical benefithas also been observed in patients with tumors where theincidence of ras mutations has historically been very low.This clearly suggests that a second important clinical phe-nomenon is underway in promoting virus efficacy. As dis-cussed extensively earlier in this review, evidence suggeststhat reovirus, similar to other viruses, manipulates theimmune system tomount an anticancer response. As intrigu-ing as this fact is, it only further affirms that the translation ofscience from "bench" to "bedside" is challenging, to say theleast.Further characterization of the activity of the virus is

clearly required and it is imperative that it be in the form ofa concerted effort of laboratory scientists with an interestin tumor biology, virologists, physician scientists, andacademic clinicians, with a common sense of purpose. Itis also critical that the patient be entirely integrated intothis effort. The availability of tumor tissue to study both,the effect of this therapy and the biology behind the drivingforce of the cancer is absolutely essential and cannot beoverstated.

It is important to note here that using viruses to targetcancer is not a new phenomenon. In fact, viral approaches tocancer have been attempted for over half a century with littlesuccess (61, 62). One of the most extensively studiedproducts is ONYX-015, an attenuated E1B-55K chimerichuman group C adenovirus, which preferentially replicateswithin and lyses tumor cells that are p53 negative (12).Almost a decade later, another novel adenovirus mutant,ONYX-053, was created that showed that loss of E1B-55K–mediated late viral RNA export, rather than p53 degrada-tion, restricts ONYX-015 replication in primary cells. It wasexperimentally proven that in contrast to the initial hypoth-esis, tumor cells that supportONYX-015 replication providethe RNA export function of E1B-55K (63). There was greatenthusiasm for this approach when a similar product, thegenetically modified adenovirus H101, made by ShanghaiSunway Biotech obtained commercial approval in China inthe treatment of advanced head and neck cancer (64). Onceagain, despite the promises of early in vivo lab work suggest-ing tumor specificity, these viruses do not specifically infectcancer cells; however, they still retain some preferential cellkill for cancer cells. As of the last report, response rates wereapproximately doubled for H101 plus chemotherapy ascompared with chemotherapy alone; however, survivalinformation is unknown. Another limitation of thisapproach is that it seems to render maximum benefit whengiven as a direct ITu injection and the patient experiences afebrile response (65). Another virus in late-stage clinicaldevelopment is Oncovex, a second generation oncolyticherpes simplex virus that expresses GM-CSF (granulocytemacrophage-colony stimulating factor) and in which dele-tion of infected cell protein (ICP) 34.5 provides tumorselectivity. This has been tested as a single-agent ITu therapyand in combination with radiotherapy (66, 67). It is cur-rently in phase III clinical development to test efficacy inmalignant melanoma (ongoing trial).Other approaches using virus-mediated oncolysis have

included the use of the oncolytic adenovirus ICOVIR-5 asa treatment for malignant gliomas, PV 701, an attenuatedform of the Newcastle virus (68, 69) and the vesicularstomatitis virus (70–73). Even more intriguing, the mea-sles virus is also being evaluated as an oncolytic virus, withencouraging data in breast and ovarian cancer (74–76).The reader is referred to more detailed reviews on onco-lytic viruses (77–79). These examples of viral oncolytictherapy elucidate the multiple challenges that we face indeveloping new viral therapies for cancer, includingreovirus.Reolysin is the trade name of the therapeutic version of

human reovirus formulated and developed by OncolyticsBiotech Inc.. It is a translucent light blue liquid containing apurified isolate of 1� 1011 TCID50 (tissue culture infectivedose) of replication competent reovirus serotype 3 Dearingstrain per milliliter in a phosphate-buffered solution and isused in many of the clinical trials. The virus in its clinicalformulation as Reolysin has rapidly progressed throughclinical development to a phase III trial in platinum-refrac-tory HNSCC (ongoing trial). An interesting pathway of the

Maitra et al.

Mol Cancer Res; 10(12) December 2012 Molecular Cancer Research1522

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 10: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

clinical development of reovirus lies in metastatic CRC;where patients, whose tumors harbor a mutation in the Krasoncogene, are ineligible to receive the anti-EGFR monoclo-nal antibodies, cetuximab and panitumumab (80, 81). Onthe basis of preclinical in vivo data that the combination ofreovirus and irinotecan is particularly synergistic in the ras-mutant cancer cells (82), the phase I study of the combi-nation of FOLFIRI (folinic acid, 5-fluorouracil, and irino-tecan) with reovirus is currently targeting patients with aKras mutation, with the potential to fulfill an unmet medi-cal need. As mentioned earlier, preclinical observations ofpreferential viral tropism under specific mutational status isnot always replicable in human subjects. The targeting ofreoviral therapy on Kras-mutated mCRC subjects is notselected on the basis of the preclinical promises of enhancedvirulence under Kras conditions but rather due to lack ofany U.S. Food and Drug Administration (FDA)-approvedtherapy for platinum-refractory mCRC subset of patients.This clearly exemplifies the future of drug development:

the potential of a safe and effective drug whose developmentis biomarker driven. The current climate of research and

health care in the United States is at a crossroads with majorchanges expected in the manner that health care is deliveredby caregivers, received by patients, and paid for by third partypayers, including the governments and the private insuranceplans. The success or failure of a drug being tested in clinicwill be highly dependent on its absolute effectiveness in anappropriate patient profile based on a validated biomarker.This is clearly a "win win" situation for all the partiesinvolved: the patient only receives the medication that ishighly likely to benefit him/her, thereby also avoidingunnecessary toxicities, and will also bring down the cost ofhealthcare by paying only for effective therapies. A word ofcaution is appropriate here: despite the encouraging devel-opment of reovirus so far, the "proof of the pudding is in theeating", and unless it can show improvement and patientbenefit over the current standard of care in a well-conductedphase III trial, it will be relegated to the confines of history, ashave many of its predecessors.

Received March 15, 2012; revised July 23, 2012; accepted September 20, 2012;published OnlineFirst October 4, 2012.

References1. Sabin AB. Reoviruses. A new group of respiratory and enteric viruses

formerly classified as ECHO type 10 is described. Science 1959;130:1387–9.

2. Nelson NJ. Viruses and cancer. J Natl Cancer Inst 1999;91:1709.3. MinHJ, Koh SS, Cho IR, Srisuttee R, Park EH, Jhun BH, et al. Inhibition

of GSK-3beta enhances reovirus-induced apoptosis in colon cancercells. Int J Oncol 2009;35:617–24.

4. WilcoxME, YangW, Senger D, Rewcastle NB,Morris DG, Brasher PM,et al. Reovirus as an oncolytic agent against experimental humanmalignant gliomas. J Natl Cancer Inst 2001;93:903–12.

5. Van DenWollenberg DJ, Van Den Hengel SK, Dautzenberg IJ, Kranen-burg O, Hoeben RC. Modification of mammalian reoviruses for use asoncolytic agents. Expert Opin Biol Ther 2009;9:1509–20.

6. Norman KL, Coffey MC, Hirasawa K, Demetrick DJ, Nishikawa SG,DiFrancesco LM, et al. Reovirus oncolysis of human breast cancer.Hum Gene Ther 2002;13:641–52.

7. Shmulevitz M, Marcato P, Lee PW. Unshackling the links betweenreovirus oncolysis, Ras signaling, translational control and cancer.Oncogene 2005;24:7720–8.

8. CoffeyMC,StrongJE, ForsythPA, LeePW.Reovirus therapyof tumorswith activated Ras pathway. Science 1998;282:1332–4.

9. Mundschau LJ, Faller DV. Endogenous inhibitors of the dsRNA-dependent eIF-2 alpha protein kinase PKR in normal and ras-trans-formed cells. Biochimie 1994;76:792–800.

10. Hata Y, Etoh T, Inomata M, Shiraishi N, Nishizono A, Kitano S, et al.Efficacy of oncolytic reovirus against humanbreast cancer cells. OncolRep 2008;19:1395–8.

11. Errington F, White CL, Twigger KR, Rose A, Scott K, Steele L, et al.Inflammatory tumour cell killing by oncolytic reovirus for the treatmentof melanoma. Gene Ther 2008;15:1257–70.

12. Bischoff JR, Kirn DH, Williams A, Heise C, Horn S, Muna M, et al. Anadenovirus mutant that replicates selectively in p53-deficient humantumor cells. Science 1996;274:373–6.

13. Errington F, Steele L, Prestwich R, Harrington KJ, Pandha HS, Vidal L,et al. Reovirus activates human dendritic cells to promote innateantitumor immunity. J Immunol 2008;180:6018–26.

14. Zitvogel L. Dendritic and natural killer cells cooperate in the control/switch of innate immunity. J Exp Med 2002;195:F9–14.

15. Hirasawa K, Nishikawa SG, Norman KL, Coffey MC, Thompson BG,Yoon CS, et al. Systemic reovirus therapy of metastatic cancer inimmune-competent mice. Cancer Res 2003;63:348–53.

16. Adair RA, Roulstone V, Scott KJ, Morgan R, Nuovo GJ, Fuller M, et al.Cell carriage, delivery, and selective replication of an oncolytic virus intumor in patients. Sci Transl Med 2012;4:138ra77.

17. Strong JE,CoffeyMC, TangD, Sabinin P, LeePW. Themolecular basisof viral oncolysis: usurpation of the Ras signaling pathway by reovirus.EMBO J 1998;17:3351–62.

18. Bischoff JR, Samuel CE. Mechanism of interferon action. Activation ofthe human P1/eIF-2 alpha protein kinase by individual reovirus s-classmRNAs: s1 mRNA is a potent activator relative to s4 mRNA. Virology1989;172:106–15.

19. Park EH, Park EH, Cho IR, Srisuttee R, Min HJ, Oh MJ, et al. CUG2, anovel oncogene confers reoviral replication through Ras and p38signaling pathway. Cancer Gene Ther 2010;17:307–14.

20. Clarke P,Meintzer SM,GibsonS,WidmannC,Garrington TP, JohnsonGL, et al. Reovirus-induced apoptosis is mediated by TRAIL. J Virol2000;74:8135–9.

21. Twigger K, Vidal L, White CL, De Bono JS, Bhide S, Coffey M, et al.Enhanced in vitro and in vivo cytotoxicity of combined reovirus andradiotherapy. Clin Cancer Res 2008;14:912–23.

22. Barkett M, Gilmore TD. Control of apoptosis by Rel/NF-kappaB tran-scription factors. Oncogene 1999;18:6910–24.

23. Strong JE, Lee PW. The v-erbB oncogene confers enhanced cellularsusceptibility to reovirus infection. J Virol 1996;70:612–6.

24. ThirukkumaranCM, Shi ZQ, Luider J, Kopciuk K, GaoH, Bahlis N, et al.Reovirus as a viable therapeutic option for the treatment of multiplemyeloma. Clin Cancer Res 2012;18:4962–72.

25. GidwaniP, ZhangW,LiuL, et al. Radiation in combinationwithReolysinfor pediatric sarcoma [Abstract]. In: Proceedings of the 99th AnnualMeeting of the American Association for Cancer Research; 2008 Apr12–16; San Diego, CA. Philadelphia, PA: AACR; 2008 Poster #3751.

26. Wadler S, Yu B, Lane M, Klampfer L, Sasazuki T, Shirasawa S, et al.The oncolytic reovirus, Reolysin augments the anticancer effects ofcytotoxic agents in vitro against the ras-mutated human colon cancercell line HCT116. European Journal of Cancer Supplements.2004;8:135.

27. Lane M, Fahey J, Besanceney C, Hamel N, Yu B, Thompson B, et al.In vivo synergy between oncolytic reovirus and gemcitabine in ras-mutated humanHCT-116 xenografts. [abstract]. In: Proceedingsof the98th Annual Meeting of the American Association for CancerResearch; 2007 Apr 14–18; Los Angeles, CA. Philadelphia, PA: AACR;2007. Abstract nr. 4812.

Development of Reovirus for Cancer Therapy

www.aacrjournals.org Mol Cancer Res; 10(12) December 2012 1523

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 11: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

28. Sei S, Mussio JK, Yang QE, Nagashima K, Parchment RE, Coffey MC,et al. Synergistic antitumor activity of oncolytic reovirus and chemo-therapeutic agents in non-small cell lung cancer cells. Mol Cancer2009;8:47.

29. Pandha HS, Heinemann L, Simpson GR, Melcher A, Prestwich R,Errington F, et al. Synergistic effects of oncolytic reovirus and cis-platin chemotherapy in murine malignant melanoma. Clin Cancer Res2009;15:6158–66.

30. Gidwani P. Systemic administration of Reolysin inhibits growthof human sarcoma xenografts alone and in combination with cis-platin and radiation. Collective Tissue Oncology Society 2008;Poster # 35039.

31. Heinemann L, Simpson GR,Boxall A, Kottke T, Relph KL,Vile R, et al.Synergistic effects of oncolytic reovirus and docetaxel chemotherapyin prostate cancer. BMC Cancer 2011;11:221.

32. Hashiro G, Loh PC, Yau JT. The preferential cytotoxicity of reovirus forcertain transformed cell lines. Arch Virol 1977;54:307–15.

33. Morris DG, Forsyth PA, Paterson AH, Fonseca K, Difrancesco LM,Thompson BG, et al. A phase I clinical trial evaluating intralesionalReolysin (reovirus) in histologically confirmed malignancies. Proc AmSoc Clin Oncol 21: 2002 (abstr 92)

34. Thirukkumaran CM, Nodwell MJ, Hirasawa K, Shi ZQ, Diaz R, LuiderJ, et al. Oncolytic viral therapy for prostate cancer: efficacyof reovirus as a biological therapeutic. Cancer Res 2010;70:2435–44.

35. Markert JM, Medlock MD, Rabkin SD, Gillespie GY, Todo T, HunterWD, et al. Conditionally replicating herpes simplex virus mutant, G207for the treatment of malignant glioma: results of a phase I trial. GeneTher 2000;7:867–74.

36. Rampling R, Cruickshank G, Papanastassiou V, Nicoll J, Hadley D,Brennan D, et al. Toxicity evaluation of replication-competent herpessimplex virus (ICP 34.5 null mutant 1716) in patients with recurrentmalignant glioma. Gene Ther 2000;7:859–66.

37. Papanastassiou V, Rampling R, Fraser M, Petty R, Hadley D, Nicoll J,et al. The potential for efficacy of the modified (ICP 34.5(-)) herpessimplex virus HSV1716 following intratumoural injection into humanmalignant glioma: a proof of principle study. Gene Ther 2002;9:398–406.

38. Harrow S, Papanastassiou V, Harland J, Mabbs R, Petty R, Fraser M,et al. HSV1716 injection into the brain adjacent to tumour followingsurgical resection of high-grade glioma: safety data and long-termsurvival. Gene Ther 2004;11:1648–58.

39. Harrington KJ, Karapanagiotou EM, Roulstone V, Twigger KR,White CL, Vidal L, et al. Two-stage phase I dose-escalation studyof intratumoral reovirus type 3 dearing and palliative radiotherapyin patients with advanced cancers. Clin Cancer Res 2010;16:3067–77.

40. Saunders M, Anthoney A, Coffey M, Mettinger K, Thompson B,Melcher A, et al. Results of a phase II study to evaluate the biologicaleffects of intratumoral (ITu) reolysin in combination with low doseradiotherapy (RT) in patients (Pts) with advanced cancers. J Clin Oncol27, 2009 (suppl; abstr e14514)

41. Gollamudi R, Ghalib MH, Desai KK, Chaudhary I, Wong B, Einstein M,et al. Intravenous administration of Reolysin, a live replication com-petent RNA virus is safe in patients with advanced solid tumors. InvestNew Drugs 2010;28:641–9.

42. Bos JL. ras oncogenes in human cancer: a review. Cancer Res1989;49:4682–9.

43. Ogino S, Kawasaki T, Brahmandam M, Yan L, Cantor M, Namgyal C,et al. Sensitive sequencing method for KRAS mutation detection byPyrosequencing. J Mol Diagn 2005;7:413–21.

44. Vidal L, Pandha HS, Yap TA, White CL, Twigger K, Vile RG, et al. Aphase I study of intravenous oncolytic reovirus type 3 Dearingin patients with advanced cancer. Clin Cancer Res 2008;14:7127–37.

45. White CL, Twigger KR, Vidal L, De Bono JS, Coffey M, Heinemann L,et al. Characterization of the adaptive and innate immune response tointravenous oncolytic reovirus (Dearing type 3) during a phase I clinicaltrial. Gene Ther 2008;15:911–20.

46. Heinemann L, Simpson GR, Boxall A, Kottke T, Relph KL, Vile R, et al.Synergistic effects of oncolytic reovirus and docetaxel chemotherapyin prostate cancer. BMC Cancer 2011;11:221.

47. HarringtonKJ, Vile RG,Melcher A, Chester J, PandhaHS. Clinical trialswith oncolytic reovirus: moving beyond phase I into combinationswithstandard therapeutics. Cytokine Growth Factor Rev 2010;21:91–8.

48. Lolkema MP, Arkenau HT, Harrington K, Roxburgh P, Morrison R,Roulstone V, et al. A phase I study of the combination of intravenousreovirus type 3 Dearing and gemcitabine in patients with advancedcancer. Clin Cancer Res 2011;17:581–8.

49. Nowak AK, Robinson BW, Lake RA. Gemcitabine exerts a selectiveeffect on the humoral immune response: implications for combinationchemo-immunotherapy. Cancer Res 2002;62:2353–8.

50. PrestwichRJ, ErringtonF, Ilett EJ,MorganRS, Scott KJ, Kottke T, et al.Tumor infection by oncolytic reovirus primes adaptive antitumorimmunity. Clin Cancer Res 2008;14:7358–66.

51. Plate JM, Plate AE, Shott S, Bograd S, Harris JE. Effect of gemcitabineon immune cells in subjects with adenocarcinoma of the pancreas.Cancer Immunol Immunother 2005;54:915–25.

52. Mita M, Wang Y, Sarantopoulos J, Vemulapalli S, George B, Mita A,et al. A Study of REOLYSIN� in Combination with Gemcitabine inPatients with Advanced Pancreatic Adenocarcinoma. In: Proceedingsof the AACR-NCIEORTC International Conference on Molecular Tar-gets and Cancer Therapeutics; 2011 Nov 12–16; San Francisco, CA.Philadelphia (PA): AACR;Mol Cancer Ther 2011;10(11 Suppl):Abstractnr B55.

53. Maddox JF, Amuzie CJ, Li M, Newport SW, Sparkenbaugh E, Cuff CF,et al. Bacterial- and viral-induced inflammation increases sensitivity toacetaminophen hepatotoxicity. J Toxicol Environ Health A 2010;73:58–73.

54. Comins C, Spicer J, Protheroe A, Roulstone V, Twigger K, White CM,et al. REO-10: a phase I study of intravenous reovirus and docetaxel inpatients with advanced cancer. Clin Cancer Res 2010;16:5564–72.

55. Garnett CT, Schlom J, Hodge JW. Combination of docetaxel andrecombinant vaccine enhances T-cell responses and antitumor activ-ity: effects of docetaxel on immune enhancement. Clin Cancer Res2008;14:3536–44.

56. Karapanagiotou EM, Roulstone V, Twigger K, Ball M, TanayM, NuttingC, et al. Phase I/II trial of carboplatin and paclitaxel chemotherapy incombination with intravenous oncolytic reovirus in patients withadvanced malignancies. Clin Cancer Res 2012;18:2080–9.

57. Karnad A, Haigentz M, Miley T, Coffey M, Gill G, Mita M. A Phase IIStudy of IntravenousWild-typeReovirus (REOLYSIN�) inCombinationwith Paclitaxel Plus Carboplatin in Patients with Platinum RefractoryMetastatic and/or Recurrent Squamous Cell Carcinoma of the Headand Neck. In: Proceedings of the AACR-NCI-EORTC InternationalConference onMolecular Targets andCancer Therapeutics; 2011 Nov12–16; San Francisco, CA. Philadelphia (PA): AACR; Mol Cancer Ther2011;10(11 Suppl):Abstract nr C22.

58. Villalona-Calero MA, Lam ET, Otterson GA, Donthireddy KR, Thur-mond J, Hade E, et al. Phase II study of reovirus with paclitaxel andcarboplatin in patients with metastatic non-small cell lung cancer(NSCLC) who have Kras or EGFR-activated tumors. 14th World LungCancer Congress, (suppl; abstr MO15.08) 2011.

59. Qiao J, Wang H, Kottke T, White C, Twigger K, Diaz RM, et al. Cyclo-phosphamide facilitates antitumor efficacy against subcutaneoustumors following intravenous delivery of reovirus. Clin Cancer Res.2008;14:259–69.

60. Suzuki E, Kapoor V, Jassar AS, Kaiser LR, Albelda SM. Gemcitabineselectively eliminates splenic Gr-1þ/CD11b þmyeloid suppressorcells in tumor-bearing animals and enhances antitumor immune activ-ity. Clin Cancer Res 2005;11:6713–21.

61. Liu TC,Galanis E,KirnD.Clinical trial resultswith oncolytic virotherapy:a century of promise, a decade of progress. Nat Clin Pract Oncol2007;4:101–17.

62. Csatary LK. Viruses in the treatment of cancer. Lancet 1971;2:825.63. O'SheaCC, JohnsonL,BagusB,Choi S,NicholasC,ShenA, et al. Late

viral RNA export, rather than p53 inactivation, determines ONYX-015tumor selectivity. Cancer cell 2004;6:611–23.

Maitra et al.

Mol Cancer Res; 10(12) December 2012 Molecular Cancer Research1524

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 12: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

64. Garber K. China approves world's first oncolytic virus therapy forcancer treatment. J Natl Cancer Inst 2006;98:298–300.

65. Xia ZJ, Chang JH, Zhang L, JiangWQ,Guan ZZ, Liu JW, et al. [Phase IIIrandomized clinical trial of intratumoral injection of E1B gene-deletedadenovirus (H101) combined with cisplatin-based chemotherapy intreating squamous cell cancer of head and neck or esophagus]. AiZheng 2004;23:1666–70.

66. Coffin RS, Hu JC, Davis CJ, Graham NJ, Groves N, Guest PJ, et al.Results of a Phase I/II clinical trial with OncoVEXGM-CSF, a secondgeneration oncolytic herpes simplex virus. J Clin Oncol 2005;23, No.16S, Part I of II (June 1 Supplement), 3099.

67. Harrington K, Hingorani M, Tanay M, Hickey J, Bhide SA, Clarke PM,et al. Phase I/II dose escalation study of OncoVexGM-CSF andchemoradiotherapy (CRT) in untreated stage III/IV squamous cellcancer of the head and neck (SCCHN). J Clin Oncol 27:15s, 2009(suppl; abstr 6018).

68. Alonso MM, Gomez-Manzano C, Jiang H, Bekele NB, Piao Y, YungWK, et al. Combination of the oncolytic adenovirus ICOVIR-5 withchemotherapy provides enhanced anti-glioma effect in vivo. CancerGene Ther 2007;14:756–61.

69. Pecora AL, Rizvi N, Cohen GI, Meropol NJ, Sterman D, Marshall JL,et al. Phase I trial of intravenous administration of PV701, an oncolyticvirus, in patients with advanced solid cancers. J Clin Oncol 2002;20:2251–66.

70. Giedlin MA, Cook DN, Dubensky TW Jr. Vesicular stomatitis virus:an exciting new therapeutic oncolytic virus candidate for cancer orjust another chapter from Field's Virology? Cancer Cell 2003;4:241–3.

71. Stojdl DF, Lichty BD, tenOever BR, Paterson JM, Power AT, KnowlesS, et al. VSV strains with defects in their ability to shutdown innateimmunity are potent systemic anti-cancer agents. Cancer Cell 2003;4:263–75.

72. Stewart JH IV, Ahmed M, Northrup SA, Willingham M, Lyles DS.Vesicular stomatitis virus as a treatment for colorectal cancer. CancerGene Ther 2011;18:837–49.

73. Bridle BW, Boudreau JE, Lichty BD, Brunelli�ere J, Stephenson K,Koshy S, et al. Vesicular stomatitis virus as a novel cancer vaccinevector to prime antitumor immunity amenable to rapid boosting withadenovirus. Mol Ther 2009;17:1814–21.

74. Hasegawa K, Nakamura T, Harvey M, Ikeda Y, Oberg A, Figini M,et al. The use of a tropism-modified measles virus in folate receptor-targeted virotherapy of ovarian cancer. Clin Cancer Res 2006;12:6170–8.

75. Iankov ID, Msaouel P, Allen C, Federspiel MJ, Bulur PA, Dietz AB, et al.Demonstration of anti-tumor activity of oncolytic measles virus strainsin amalignant pleural effusion breast cancermodel. Breast Cancer ResTreat 2010;122:745–54.

76. Russell SJ, Peng KW. Measles virus for cancer therapy. Curr TopMicrobiol Immunol 2009;330:213–41.

77. Bell J. Oncolytic viruses: an approved product on the horizon? MolTher 2010;18:233–4.

78. Wong HH, Lemoine NR, Wang Y. Oncolytic Viruses for Cancer Ther-apy: Overcoming the Obstacles. Viruses 2010;2:78–106.

79. Zeyaullah M, Patro M, Ahmad I, Ibraheem K, Sultan P, Nehal M, et al.Oncolytic viruses in the treatment of cancer: a review of currentstrategies. Pathol Oncol Res 2012;18:771–81.

80. Amado RG, Wolf M, Peeters M, Van Cutsem E, Siena S, FreemanDJ, et al. Wild-type KRAS is required for panitumumab efficacy inpatients with metastatic colorectal cancer. J Clin Oncol 2008;26:1626–34.

81. DeRoockW, PiessevauxH, De Schutter J, JanssensM,DeHertoghG,Personeni N, et al. KRAS wild-type state predicts survival and isassociated to early radiological response in metastatic colorectalcancer treated with cetuximab. Ann Oncol 2008;19:508–15.

82. Seetharam R, Coffey M, Klampfer L, Mariadason J, Goel S. Theaddition of Reolysin, an oncolytic reovirus, to irinotecan shows syn-ergistic anti cancer activity in colorectal cancer cell lines [Abstract]. In:Proceedings of the 101st Annual Meeting of the American Associationof Cancer Research, 2010 Mar 31–April 3; Washington, DC. Philadel-phia (PA): AACR; 2010. Abstract nr # 1773.

www.aacrjournals.org Mol Cancer Res; 10(12) December 2012 1525

Development of Reovirus for Cancer Therapy

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157

Page 13: Reovirus: A Targeted Therapeutic Progress And Potential · dsRNA (5). However, considering the fact that this naturally oncolytic, and pathologically self resolving virus seems to

2012;10:1514-1525. Published OnlineFirst October 4, 2012.Mol Cancer Res   Radhashree Maitra, Mohammad H. Ghalib and Sanjay Goel 

Progress And Potential−−Reovirus: A Targeted Therapeutic

  Updated version

  10.1158/1541-7786.MCR-12-0157doi:

Access the most recent version of this article at:

   

   

  Cited articles

  http://mcr.aacrjournals.org/content/10/12/1514.full#ref-list-1

This article cites 72 articles, 28 of which you can access for free at:

  Citing articles

  http://mcr.aacrjournals.org/content/10/12/1514.full#related-urls

This article has been cited by 7 HighWire-hosted articles. Access the articles at:

   

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

  Subscriptions

Reprints and

  [email protected]

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

  Permissions

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

.http://mcr.aacrjournals.org/content/10/12/1514To request permission to re-use all or part of this article, use this link

on May 15, 2021. © 2012 American Association for Cancer Research. mcr.aacrjournals.org Downloaded from

Published OnlineFirst October 4, 2012; DOI: 10.1158/1541-7786.MCR-12-0157