11
Translational Cancer Mechanisms and Therapy Ad5 NULL -A20: A Tropism-Modied, avb6 Integrin-Selective Oncolytic Adenovirus for Epithelial Ovarian Cancer Therapies Hanni Uusi-Kerttula 1 , James A. Davies 1 , Jill M. Thompson 2 , Phonphimon Wongthida 2 , Laura Evgin 2 , Kevin G. Shim 2 , Angela Bradshaw 3 , Alexander T. Baker 1 , Pierre J. Rizkallah 4 , Rachel Jones 5 , Louise Hanna 6 , Emma Hudson 6 , Richard G. Vile 2 , John D. Chester 1,6 , and Alan L. Parker 1 Abstract Purpose: Virotherapies are maturing in the clinical setting. Adenoviruses (Ad) are excellent vectors for the manipulability and tolerance of transgenes. Poor tumor selectivity, off-target sequestration, and immune inactivation hamper clinical efcacy. We sought to completely redesign Ad5 into a rened, tumor-selective virotherapy targeted to avb6 integrin, which is expressed in a range of aggressively transformed epithelial cancers but nondetectable in healthy tissues. Experimental Design: Ad5 NULL -A20 harbors mutations in each major capsid protein to preclude uptake via all native pathways. Tumor-tropism via avb6 targeting was achieved by genetic insertion of A20 peptide (NAVPNLRGDLQVLAQK- VART) within the ber knob protein. The vector's selectivity in vitro and in vivo was assessed. Results: The tropism-ablating triple mutation completely blocked all native cell entry pathways of Ad5 NULL -A20 via coxsackie and adenovirus receptor (CAR), avb3/5 integrins, and coagulation factor 10 (FX). Ad5 NULL -A20 efciently and selectively transduced avb6 þ cell lines and primary clinical ascites-derived EOC ex vivo, including in the presence of preexisting anti-Ad5 immunity. In vivo biodistribution of Ad5 NULL -A20 following systemic delivery in nontumor- bearing mice was signicantly reduced in all off-target organs, including a remarkable 10 7 -fold reduced genome accumulation in the liver compared with Ad5. Tumor up- take, transgene expression, and efcacy were conrmed in a peritoneal SKOV3 xenograft model of human EOC, where oncolytic Ad5 NULL -A20treated animals demonstrated sig- nicantly improved survival compared with those treated with oncolytic Ad5. Conclusions: Oncolytic Ad5 NULL -A20 virotherapies rep- resent an excellent vector for local and systemic targeting of avb6-overexpressing cancers and exciting platforms for tumor-selective overexpression of therapeutic anticancer modalities, including immune checkpoint inhibitors. Clin Cancer Res; 24(17); 421524. Ó2018 AACR. Introduction Ovarian cancer remains the deadliest gynecologic cancer with global 5-year survival rates below 50% (1). The early stages of the disease are commonly asymptomatic, with the result that most patients have advanced, incurable disease, at presenta- tion. Ovarian cancer metastasizes with large volumes of malig- nant, intraperitoneal ovarian ascites (OAS) providing a protu- morigenic microenvironment (2). Chemoresistance rapidly develops during treatment, requiring alternative regimens. Epithelial ovarian cancer (EOC) is the most common (90%) ovarian cancer type (3). One-third of patients with EOC have cells expressing an epithelial cancer-specic marker, avb6 integ- rin (4). Upregulation of avb6 expression in cancer has been linked to aggressive transformation, metastasis, and poor prog- nosis (58). avb6 is absent in healthy epithelium (5, 9) but widely overexpressed in plethora of cancers, including ovarian, lung, skin, esophageal, cervical, and head and neck cancers (4), thus making it a promising target for therapeutic vectors. avb6 is an activator of TGFb1 signaling that promotes metastasis by enhancing angiogenesis, immune cell suppression, and epithe- lial-to-mesenchymal transition (reviewed in ref. 10). Cancer virotherapy is undergoing renewed interest, includ- ing recent regulatory approval for clinical use of herpes simplex type 1based talimogene laherparepvec (T-VEC), the rst oncolytic immunotherapy approved for advanced mela- noma (11). Very recently, oncolytic viruses were shown to sensitize difcult-to-treat tumors, including triple-negative breast cancer (TNBC; ref. 12) and glioblastoma (13) to subse- quent immunotherapies with immune checkpoint inhibitors. This highlights the potential of virotherapies for combination 1 Division of Cancer and Genetics, Cardiff University, Cardiff, United Kingdom. 2 Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota. 3 BHF Glasgow Cardiovascular Research Centre, Glasgow, United Kingdom. 4 Division of Infection and Immunity, Cardiff University, Cardiff, United Kingdom. 5 South West Wales Cancer Institute, Singleton Hospital, Swansea, United Kingdom. 6 Velindre Cancer Centre, Cardiff, United Kingdom. Note: Supplementary data for this article are available at Clinical Cancer Research Online (http://clincancerres.aacrjournals.org/). Corresponding Author: Alan L. Parker, Division of Cancer and Genetics, Henry Wellcome Building, Cardiff University School of Medicine, Heath Park, Cardiff CF14 4XN, United Kingdom. Phone: 44-2922510231; Fax: 44-2920687006; E-mail: [email protected] doi: 10.1158/1078-0432.CCR-18-1089 Ó2018 American Association for Cancer Research. Clinical Cancer Research www.aacrjournals.org 4215 on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

Ad5 -A20: A Tropism-Modified, avb6 Integrin-Selective ... · wild-type Ad5 for tumor therapy. We have generated a novel virotherapy vector, Ad5 NULL-A20, with altered, tumor-selective

  • Upload
    others

  • View
    3

  • Download
    0

Embed Size (px)

Citation preview

  • Translational Cancer Mechanisms and Therapy

    Ad5NULL-A20: A Tropism-Modified, avb6Integrin-Selective Oncolytic Adenovirus forEpithelial Ovarian Cancer TherapiesHanni Uusi-Kerttula1, James A. Davies1, Jill M. Thompson2,Phonphimon Wongthida2, Laura Evgin2, Kevin G. Shim2, Angela Bradshaw3,Alexander T. Baker1, Pierre J. Rizkallah4, Rachel Jones5, Louise Hanna6,Emma Hudson6, Richard G. Vile2, John D. Chester1,6, and Alan L. Parker1

    Abstract

    Purpose: Virotherapies are maturing in the clinical setting.Adenoviruses (Ad) are excellent vectors for the manipulabilityand tolerance of transgenes. Poor tumor selectivity, off-targetsequestration, and immune inactivation hamper clinicalefficacy. We sought to completely redesign Ad5 into a refined,tumor-selective virotherapy targeted to avb6 integrin, whichis expressed in a range of aggressively transformed epithelialcancers but nondetectable in healthy tissues.

    Experimental Design: Ad5NULL-A20 harbors mutations ineach major capsid protein to preclude uptake via all nativepathways. Tumor-tropism via avb6 targeting was achieved bygenetic insertion of A20 peptide (NAVPNLRGDLQVLAQK-VART) within the fiber knob protein. The vector's selectivityin vitro and in vivo was assessed.

    Results: The tropism-ablating triple mutation completelyblocked all native cell entry pathways of Ad5NULL-A20 viacoxsackie and adenovirus receptor (CAR), avb3/5 integrins,and coagulation factor 10 (FX). Ad5NULL-A20 efficiently and

    selectively transduced avb6þ cell lines and primary clinicalascites-derived EOC ex vivo, including in the presence ofpreexisting anti-Ad5 immunity. In vivo biodistribution ofAd5NULL-A20 following systemic delivery in non–tumor-bearing mice was significantly reduced in all off-targetorgans, including a remarkable 107-fold reduced genomeaccumulation in the liver compared with Ad5. Tumor up-take, transgene expression, and efficacy were confirmed in aperitoneal SKOV3 xenograft model of human EOC, whereoncolytic Ad5NULL-A20–treated animals demonstrated sig-nificantly improved survival compared with those treatedwith oncolytic Ad5.

    Conclusions: Oncolytic Ad5NULL-A20 virotherapies rep-resent an excellent vector for local and systemic targetingof avb6-overexpressing cancers and exciting platformsfor tumor-selective overexpression of therapeutic anticancermodalities, including immune checkpoint inhibitors.Clin Cancer Res; 24(17); 4215–24. �2018 AACR.

    IntroductionOvarian cancer remains the deadliest gynecologic cancer with

    global 5-year survival rates below 50% (1). The early stages ofthe disease are commonly asymptomatic, with the result thatmost patients have advanced, incurable disease, at presenta-tion. Ovarian cancer metastasizes with large volumes of malig-nant, intraperitoneal ovarian ascites (OAS) providing a protu-

    morigenic microenvironment (2). Chemoresistance rapidlydevelops during treatment, requiring alternative regimens.Epithelial ovarian cancer (EOC) is the most common (90%)ovarian cancer type (3). One-third of patients with EOC havecells expressing an epithelial cancer-specific marker, avb6 integ-rin (4). Upregulation of avb6 expression in cancer has beenlinked to aggressive transformation, metastasis, and poor prog-nosis (5–8). avb6 is absent in healthy epithelium (5, 9) butwidely overexpressed in plethora of cancers, including ovarian,lung, skin, esophageal, cervical, and head and neck cancers (4),thus making it a promising target for therapeutic vectors. avb6is an activator of TGFb1 signaling that promotes metastasis byenhancing angiogenesis, immune cell suppression, and epithe-lial-to-mesenchymal transition (reviewed in ref. 10).

    Cancer virotherapy is undergoing renewed interest, includ-ing recent regulatory approval for clinical use of herpessimplex type 1–based talimogene laherparepvec (T-VEC), thefirst oncolytic immunotherapy approved for advanced mela-noma (11). Very recently, oncolytic viruses were shown tosensitize difficult-to-treat tumors, including triple-negativebreast cancer (TNBC; ref. 12) and glioblastoma (13) to subse-quent immunotherapies with immune checkpoint inhibitors.This highlights the potential of virotherapies for combination

    1Division of Cancer and Genetics, Cardiff University, Cardiff, United Kingdom.2Department of Molecular Medicine, Mayo Clinic, Rochester, Minnesota. 3BHFGlasgow Cardiovascular Research Centre, Glasgow, United Kingdom. 4Divisionof Infection and Immunity, Cardiff University, Cardiff, United Kingdom. 5SouthWest Wales Cancer Institute, Singleton Hospital, Swansea, United Kingdom.6Velindre Cancer Centre, Cardiff, United Kingdom.

    Note: Supplementary data for this article are available at Clinical CancerResearch Online (http://clincancerres.aacrjournals.org/).

    Corresponding Author: Alan L. Parker, Division of Cancer and Genetics, HenryWellcome Building, Cardiff University School of Medicine, Heath Park, CardiffCF14 4XN, United Kingdom. Phone: 44-2922510231; Fax: 44-2920687006;E-mail: [email protected]

    doi: 10.1158/1078-0432.CCR-18-1089

    �2018 American Association for Cancer Research.

    ClinicalCancerResearch

    www.aacrjournals.org 4215

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://crossmark.crossref.org/dialog/?doi=10.1158/1078-0432.CCR-18-1089&domain=pdf&date_stamp=2018-8-9http://clincancerres.aacrjournals.org/

  • studies in the clinical setting, and the scope for generating avector capable of systemically targeting tumors following intra-venous introduction. Adenovirus serotype 5 (Ad5) has beencommonly deployed in clinical trials of cancer and gene ther-apies (14), due to ease of genetic manipulation and capacity forlarge transgenes (15). However, this serotype has suboptimalfeatures that hamper its wider clinical use. As a commonrespiratory virus with high seroprevalence rates (16), efficientneutralization of vector by neutralizing antibodies (nAb) limitsefficacy. Other limitations include significant and rapid off-target sequestration to spleen and liver via complexing of thevirion with human coagulation factor 10 (FX; ref. 17) andpotentially other coagulation factors (reviewed extensively inref. 18), "bridging" the complex to heparan sulphate proteo-glycans (HSPG), abundant on hepatocytes (19). In vitro, Ad5enters host cells via coxsackie and adenovirus receptor (CAR;ref. 20) that is ubiquitous within tight junctions on polarizedepithelial cells (reviewed in ref. 21) but commonly down-regulated in progressive cancers (22–26), limiting use ofwild-type Ad5 for tumor therapy.

    We have generated a novel virotherapy vector, Ad5NULL-A20,with altered, tumor-selective tropism. We ablated all nativetropisms of Ad5 by mutating key residues in the three maincapsid proteins (hexon, fiber, and penton) and retargeted theresulting vector, Ad5NULL, to the tumor-selective integrin avb6through incorporation of an avb6-binding peptide (A20,NAVPNLRGDLQVLAQKVART) within the fiber knob domain HIloop, generating the novel vector Ad5NULL-A20. A20 peptide wasoriginally derived from foot-and-mouth disease virus (FMDV)capsid protein VP1, and has high affinity for its native receptor,avb6 integrin (27, 28). We have investigated potential clinicalutility of an oncolytic variant of Ad5NULL-A20 (D24/T1) forintraperitoneal treatment of ovarian cancer by investigatingits biodistribution, tumor-selective oncolytic capabilities andavoidance of immune neutralization using in vitro and in vivomodels of human EOC.

    Materials and MethodsAdenovirus vectors, cell lines, and clinical ascites

    All vectors generated in this study included a luciferase (Luc)reporter gene. Genetic modifications were carried out by AdZhomologous recombineering methods (29) as described pre-viously (30). Viruses were produced in T-REx-293 or HEK293-b6 cells (for A20-modified viruses) and purified as describedpreviously (30, 31). A triply detargeted vector genome,Ad5NULL, was generated by introducing mutations in key genesencoding of each of the major capsid proteins to precludecellular uptake by all known native Ad5 pathways. Ablationof binding to CAR was achieved via the KO1mutation in the ABloop of the L5 fiber knob gene; ablation of binding to co-agulation factor 10 (FX) via a mutation in hypervariable region7 of the L3 hexon gene; and ablation of avb3/5 integrin bindingvia RGD-to-RGE mutation in the L2 penton base gene. avb6retargeting was achieved by insertion of sequences encodingpeptide A20 (NAVPNLRGDLQVLAQKVART) into the fiberknob HI loop (between residues G546 and D547) of Ad5NULL,generating Ad5NULL-A20. Replication-deficient variants ofAd5NULL-A20 carry a complete E1/E3 deletion. Oncolytic var-iants have a 24-base pair deletion (dl922–947) in the retino-blastoma protein (pRB) binding domain of E1A (D24; ref. 32)and a single adenine insertion at position 445 within theendoplasmic reticulum (ER) retention domain of E3/19K(T1 mutation; ref. 33). Additional details of genetic modifica-tions are provided in Supplementary Methods.

    Homology modeling was performed using the previouslypublished Ad5 fiber knob structure (PDB ID: 1KNB; ref. 34) andFMDV O PanAsia VP1 protein in complex with anb6 (PDB ID:5NEM; ref. 35). The peptide sequence forming the inter-action with anb6 (NVRGDLQVLAQKVART) was edited to con-form with the A20 peptide sequence used in this study(NAVPNLRGDLQVLAQKVART), docked to the Ad5 fiber knobstructure in the HI loop and the KO1 mutation added usingWinCoot (36) and PyMol 2.0 (37). The crude Ad5NULL-A20structure was aligned with the existing 5NEM structure and thecomplex energy minimized using the YASARA algorithm (38).Binding energy calculations were performed using PISA (39),and surface charge was calculated using APBS tool in PyMol2.0 (37).

    avb6-high/CARþ SKOV3-b6 cell line was generated in-houseby retroviral transfection of SKOV3 cells (that natively express theav subunit; ref. 40) with integrin beta6 pBABE puro plasmid toexpress the b6 subunit. Primary EOC cells from ascites wereobtained through Wales Cancer Bank under existing ethicalpermissions (WCB 14/004). Cells were processed and subcul-tured as described previously (30, 31), and tested regularly forMycoplasma infection by commercially available PCR-basedmethods.

    In vitro and in vivo studiesCell surface receptor expression was assessed by flow cyto-

    metry (30). The presence of anti Ad5 antibodies in OAS andserum was determined by ELISA as previously reported (41).Antigen specificity of the antibodies was assessed by Westernblot. Transduction efficiency was assessed by standard lucifer-ase assays, described previously (30, 31). Animal experimentswere approved by Institutional Care and Use Committee(IACUC) and performed at Mayo Clinic, Rochester, MN.Animals were age and sex matched. Animal handling and

    Translational Relevance

    Virotherapies are emerging as clinically important anti-cancer agents, demonstrating synergy with immune check-point inhibitors in several recent, high-profile studies.Because these agents have not evolved to be intrinsicallytumor selective, therapeutic index could be furtherenhanced by a thorough redesign of the virus capsid toimprove tumor selectivity following intravascular delivery.To this end, we have systematically refined the adenovirusserotype 5 (Ad5) capsid to genetically preclude uptake viaall known native cellular entry pathways, to generate a basaland more biocompatible vector, Ad5NULL. To empower thisvector with tumor selectivity, we further engineered theAd5NULL capsid to present a high-affinity avb6 integrin-binding oligopeptide, A20. The resultant virotherapy,Ad5NULL-A20 demonstrates exquisite tumor selectivity bothin vitro and in vivo, with basal "off-target" uptake. Ad5NULL-A20 thus represents a powerful platform for targeted in situoverexpression of immunomodulatory modalities for futuretranslational applications.

    Uusi-Kerttula et al.

    Clin Cancer Res; 24(17) September 1, 2018 Clinical Cancer Research4216

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • injections were performed by a veterinary technologist. In vivoexperiments are further described in detail in SupplementaryMethods.

    Statistical analysesFigures and statistical analyses were generated using GraphPad

    Prism v6.03. In vitro and ex vivo assays were analyzed by two-tailed unpaired t tests or one-way ANOVA with the Dunnettmultiple-comparisons post hoc test. In vivo data were normal-ized and analyzed by one-way ANOVA or Kruskal–Wallis testwith Sidak or Dunn multiple-comparisons post hoc test, respec-tively. Overall survival (%) following oncolytic treatment isshown as a Kaplan–Meier survival curve; survival proportionswere analyzed by the Gehan–Breslow–Wilcoxon test.

    ResultsWe generated and produced to very high viral titers replication-

    defective and oncolytic variants of a novel Ad5NULL-A20 vector(Fig. 1A) with three detargeting mutations and an A20 peptideinsertion that retargets the vector toavb6 integrin-expressing cells(Fig. 1B). Additionally, we generated replication deficient andoncolytic versions of Ad5.A20, which harbors the avb6 targeting-peptide A20 insertion, in the absence of any detargeting modi-fication. The multiple genetic manipulations did not have asignificant impact on viral titer (Fig. 1A).

    We generated a homology model of the Ad5NULL-A20 fiberknob protein in complex with the anb6 dimer (Fig. 1C). The A20peptide (dark blue) occupies space spanning both an (green) and

    b6 (purple) subunits. The predicted Ad5NULL-A20 interactingresidues of the A20 peptide (dark blue) and the native knobstructure (cyan) against the approximated charge surface of theanb6 (red is negative, blue is positive; Fig. 1D). The anb6 hasmostly negative surface potential in this region (1D), comple-mentary to the predominantly positive charge of theAd5NULL-A20interface (Supplementary Fig. SI A). The adjacent CD loop of thenative Ad5 fiber knob contributes two polar residue interactionsfrom Lys-442 and Gly-443 (1D), binding to an additional threean residues (Supplementary Fig. SI B). The binding energy of theanb6-Ad5NULL-A20 fiber knob complex is calculated to be –24.3Kcal/mol, suggesting an exceptionally stable interface (Supple-mentary Fig. SI C), providing confidence that our avb6 targetingstrategy was feasible.

    The transduction efficiency of replication-deficient vectors wasassessed in cell lines expressing variable levels of CAR and avb6integrin. The detargeting mutation triplet of Ad5NULL-A20completely abolished entry via CAR in CHO-CAR cells (CARþ),while Ad5 transduced these cells at expectedly high efficiency(Fig. 2A). The HVR7mutation abolished Ad5 vector transductionvia FX pathway (Fig. 2B; ref. 42). As expected, FX significantlyincreased transduction of Ad5 into CHO-K1 cells as comparedwith FX-free culture conditions (Fig. 2B, left). Conversely, addi-tion of human FX in culture medium had no effect on thetransduction efficiency of the FX binding-ablated Ad5.HVR7control vector in these cells (Fig. 2B; right). Furthermore, theenhanced transduction seen for Ad5 was reversed by the additionof a 3:1 molar excess of Gla-domain interacting protein, antico-agulant X-bp, which binds and inactivates FX in the medium

    Figure 1.

    Generated vectors. A, Viral titers andexpected tropisms of Ad5 and triplydetargeted, avb6 integrin retargetedvector, Ad5NULL-A20;B,Vectormap ofthe oncolytic Ad5NULL-A20; C,Homology modeling of the Ad5 fiberknob with A20 peptide(NAVPNLRGDLQVLAQKVART; darkblue) within the HI loop of fiber knobdomain (Ad5.A20; in light blue) incomplex with an (green) and b6(magenta) integrin subunits shows apotential mechanism for the Ad5NULL-A20 interface. D, Residues in both anand b6 subunits form hydrogen bonds(red dashes), stabilizing a chargedinterface (anb6, negative; A20,positive). Residues in Ad5s CD loopform further polar interactions. CAR,coxsackie and adenovirus receptor;FX, coagulation factor 10; HVR7,hypervariable region 7 (42); KO1,CAR-binding mutation in fiber knobAB loop (53); Luc, luciferasetransgene; repl. def., replication-defective; vp, viral particle.

    Ad5NULL-A20: An Exquisitely Tumor-Selective Virotherapy

    www.aacrjournals.org Clin Cancer Res; 24(17) September 1, 2018 4217

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • (ref. 19; Fig. 2B, left). On the contrary, FX depletion did not affectthe transduction of Ad5.HVR7 vector (Fig. 2B, right).

    We confirmed avb6 integrin as the primary entry receptorfor the triply detargeted, integrin retargeted Ad5NULL-A20 vector(Fig. 3). Ad5NULL-A20 transduced avb6þ/CAR� BT-20 breast can-cer cells with 305-fold higher efficiency (Fig. 3A; P¼ 0.0270) andprimary, patient-derived EOC004 cells (avb6þ/CAR�) at 69-foldincreased efficiency (Fig. 3B; P ¼ 0.0090) relative to Ad5. Com-petition assays using a function-blocking anti-avb6 antibody

    (10D5) significantly inhibited cell transduction by Ad5NULL-A20 vector in SKOV3-b6 cells (avb6þ/CARþ; Fig. 3C; P ¼0.0010), confirming the vector's selectivity for avb6 integrin.

    We next evaluated the ability of the Ad5NULL-A20 vector toretain its infectivity in the highly neutralizing environmentpresented by OAS. To this end, freshly isolated clinical OASsamples from 20 patients with ovarian cancer were screenedfor the presence of anti-Ad5 antibodies by direct ELISA. Thetiters of anti-Ad5 abs in malignant OAS were scrutinized againstthe serum anti-Ad5 antibody titer of a healthy adult malevolunteer (Fig. 4A). Equal proportions of patients were foundto have lower and higher antibody titers than the control serum(Fig. 4A, black dashed line). Ascites from patient 001 (OAS001)were chosen for subsequent neutralization assays due to itssimilar antibody titer with the control serum. Antibodies inOAS001 and control serum appeared specific for the viral fiberprotein, whilst the most abundant capsid protein—hexon—was recognized only at very low levels in Western blot usingdenatured whole viral particles (Fig. 4B). The neutralizing effectof OAS001 on transduction efficiency of Ad5NULL-A20 wasassessed in avb6þ/CAR� EOC004 primary cells. Ad5NULL-A20 showed up to 902-fold higher transduction efficiency inprimary human EOC cultures relative to Ad5 at OAS concen-trations of 2.5%, 5%, and 10%, whilst Ad5 was not capable oftransducing these cells at detectable levels (Fig. 4C).

    We next evaluated biodistribution of virus infection in im-munocompetent, non–tumor-bearing mice. Mice were injectedintravenously with replication-defective vectors to assess in vivotropism (Fig. 5A), in particular the effect of the three detarget-ing mutations on biodistribution of virus infection. As expect-ed and as previously documented, the Ad5 vector showedintense localization in the area of liver and spleen, whileluminescence by the Ad5NULL-A20 vector was completely un-detectable at the 72-hour time point (Fig. 5B). Animals inoc-ulated with Ad5 vector had significantly higher whole-bodyluminescence than the control animals (P < 0.0001) or theAd5NULL-A20 vector (P < 0.0001; Fig. 5C). The liver, spleen,lungs, ovaries, and heart were resected postmortem and quan-tified for ex vivo luminescence (for luminescence heatmaps,see Supplementary Fig. SIIA–C). The livers of Ad5-challengedanimals emitted significantly more luminescence than thePBS control or Ad5NULL-A20 groups (both P < 0.0001;Fig. 5D). Similarly, Ad5NULL-A20 had significantly decreasedtransgene expression in the spleen, lungs, ovaries and heart,relative to Ad5 (Fig. 5E–H; P < 0.0001 for all). For fold changes

    Figure 2.

    Ablation of native receptor tropisms. A,Binding of replication-deficient Ad5 andAd5NULL-A20 vectors to coxsackie andadenovirus receptor (CAR). Ratio of viraltransgene expression from Ad5NULL-A20relative to Ad5 is indicated above bars. B,Binding of replication-deficient Ad5 andHVR7-mutated Ad5 variant (42) tocoagulation factor 10 (FX) was assessed inluciferase assays by infecting CHO-K1 cells inthe presence of human FX with (þ) orwithout (�) anticoagulant X-bp. HVR7, FX-bindingmutation. Statistical significance: ns,P > 0.05; �� , P < 0.01.

    Figure 3.

    In vitro assessment of avb6 integrin retargeting. Transduction efficiency ofreplication-deficient wild-type (Ad5) and triply detargeted, integrin retargeted(Ad5NULL-A20) vectors in (A) avb6þ BT-20 breast cancer cells and (B) avb6þ

    primary EOC cells from patient 004. C, Competition inhibition of avb6 integrin-mediated cell entry in SKOV3-b6 cells. The highest 10% avb6-expressingSKOV3-b6 cells were sorted by FACS, subcultured, and infected. IgG, normalmouse IgG control; 10D5, anti-avb6 function-blocking antibody. Ratio of viraltransgene expression is indicated above bars. Statistical significance: ns,P > 0.05; � , P < 0.05; �� , P < 0.01; ��� , P < 0.001; ���� , P < 0.0001.

    Uusi-Kerttula et al.

    Clin Cancer Res; 24(17) September 1, 2018 Clinical Cancer Research4218

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • in luminescence intensity in each off-target organ, see Supple-mentary Fig. SIID.

    Confirmation that the modifications in Ad5NULL-A20 result-ed in reduced sequestration of virus in multiple normaltissues was performed via quantitation of viral load by qPCR.Genome copy-number of the Ad5NULL-A20 vector was 10million times lower in the liver relative to the Ad5 (Fig. 6A;P < 0.0001). Similarly, Ad5NULL-A20 genome copy-number wasover 700-fold lower in the spleen compared with Ad5 (Fig. 6B;P < 0.0001). In addition, the Ad5NULL-A20 vector showedimproved off-target profiles in all organs relative to Ad5, withviral load 105, 104, and 103 lower in the lungs, heart, andovaries, respectively (Fig. 6C–E). Successful detargeting of theliver being due to our genetic modifications of Ad5 is support-ed by immunohistochemical staining of liver sections,which showed high expression levels of CAR, whilst avb6 was

    undetectable (Supplementary Fig. SIIIA). Confirmation of thedetargeting effects of genetic modifications in Ad5NULL-A20is provided by the observation that liver sections from miceshowed positive staining for Ad capsid proteins in the Ad5group, but not in livers of mice that had been challenged withthe Ad5NULL-A20 vector (Supplementary Fig. SIIIB).

    To evaluate efficacy in a human EOC model in vivo, SKOV3human ovarian cancer xenografts were established in immuno-compromised NOD/SCID mice. Animals developed large solidtumors at the cell injection site and at various sites within theperitoneal cavity within 14 days after intraperitoneal implanta-tion of SKOV3 cells (for tumor localization and take rate; seeSupplementary Fig. SIV) and by day 49, tumors were spreadthroughout the peritoneal cavity with accumulation of largevolumes of ascites. Based on these observations, we performedvirotherapy efficacy studies by delivering three intraperitoneal

    Figure 4.

    The effect of malignant OAS on vector transduction ex vivo. A, Quantification of anti-Ad5 antibodies in 20 clinical OAS samples and control serum from ahealthy male volunteer (solid black line) by ELISA. Horizontal lines indicate 50% and 100% binding of anti-Ad5 abs in the control serum. B, Antigenspecificity of anti-Ad5 antibodies in ascites and serum by Western blot, using denatured whole virus particles. C, Vector transduction efficiency of replication-defective (Ad5) and Ad5NULL-A20 vectors, in the absence and presence of varying dilutions of ascites from an ovarian cancer patient 004 in primary ex vivoculture of EOC cells from patient 004. Cells were preincubated with ascending concentrations of ascites and infected.

    Ad5NULL-A20: An Exquisitely Tumor-Selective Virotherapy

    www.aacrjournals.org Clin Cancer Res; 24(17) September 1, 2018 4219

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • doses of oncolytic variants of Ad5, Ad5.A20, and Ad5NULL-A20vectors on days 14, 16, and 18 after implantation of SKOV3 cells.

    IVIS imaging at 48 hours after first virotherapy treatmentdose (day 16) showed widespread luminescence throughoutthe abdominal region in animals with SKOV3 xenografts andtreated with the oncolytic Ad5 vector, with highest intensity inthe liver/spleen region (Fig. 7B). This distribution was main-tained, but at lower intensity, until 5 days later, day 21 (Fig.7B). In contrast, the oncolytic Ad5NULL-A20 vector, however,showed selective tumor localization, with significantly reducedoverall luminescence relative to Ad5, consistent with successfuldetargeting of non-tumor tissues. The distribution of infectionmediated by the oncolytic Ad5.A20 vector was intermediatebetween the Ad5 and Ad5NULL-A20. Quantitation of total bodyluminescence showed uptake of the Ad5NULL-A20 vector to besignificantly lower than Ad5 both on day 16 (Fig. 7C; P < 0.05

    and 0.05;� , P < 0.05; �� , P < 0.01; ��� , P < 0.001;���� , P < 0.0001.

    Uusi-Kerttula et al.

    Clin Cancer Res; 24(17) September 1, 2018 Clinical Cancer Research4220

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • known native tropisms and retargeted to an overexpressed, prog-nostic cancer marker—avb6 integrin (43). Integrin avb6 is apromising target for therapeutic cancer applications due to itsoverexpression in aggressively transformed cancers (4). A20 pep-tide is a feasible tool for a variety of clinical applications andhas been used for imaging diagnostics in an avb6þ pancreatictumor model (44) and in a humanized single-chain Fv antibodyB6-2 (45). avb6 is emerging as a promising target for a range ofadvances therapies, including those based on chimeric antigenreceptor (CAR) T-cell immunotherapies (reviewed in ref. 46),where efficacy in the avb6 expressing SKOV3 cell lines hasbeen demonstrated. Furthermore, the avb6-blocking antibody264RAD showed promising in vivo efficacy in HER2þ/avb6þ

    breast cancers in combination with the monoclonal antibodytrastuzumab (47) and is being developed for phase I clinicaltrials. avb6 therefore represents a highly appealing target forcancer treatment across a range of technologies and therapeuticapplications.

    In silico evaluation of the Ad5NULL-A20 interface with anb6by homology modeling (Fig. 1; Supplementary Fig. SI) predictsthe Ad5NULL-A20 fiber knob domain to form a low entropyinterface with anb6. A20 possesses the putative RGD integrininteracting motif (48), but specificity to the b6 subunit isderived from the helical motif C-terminal of RGD. It is furtherstabilized by electrostatic interactions across the interface and

    polar bonds between an and the Ad5 CD loop. Each fibertrimer possesses three copies of the A20 peptide, with 12trimeric fibers per adenovirus capsid, thus Ad5NULL-A20 pos-sesses 36 potential anb6 interaction sites per viral particle.While not all these sites will be utilized in a single cellularinteraction, it is extremely likely that the virus benefits from apotent avidity effect when interacting with a cell possessingmultiple anb6 copies.

    In the present study, we presented the Ad5NULL-A20 as ahighly selective vector platform. A replication-defective form ofAd5NULL-A20 vector successfully detargeted viral uptake by cellsvia native viral uptake pathways (Fig. 2), instead selectivelyretargeting avb6þ cells, in vitro and ex vivo (Fig. 3). Although theefficacy-limiting interactions that occur in the systemic deliveryof adenoviral vectors can, theoretically, be bypassed by intra-cavity administration of the vector via the i.p. route, in practicethis approach presents challenges because wild-type Ad5 issequestered by preexisting anti-Ad5 immunity in the form ofneutralizing antibodies (nAb) in ascitic fluid (41, 49, 50). Wetherefore assessed the transduction efficiency of Ad5NULL-A20in the presence of freshly isolated clinical OAS from patientswith ovarian cancer with confirmed high levels of anti-Ad5nAbs (Fig. 4A). Unlike the Ad5 vector, Ad5NULL-A20 retained itsability to transduce avb6þ cells, even at relatively high OASconcentrations (Fig. 4C).

    Figure 6.

    Viral genome copy-number in off-target organs at 72 hours followingsystemic delivery. Adenovirus genomecopy-number from tissues excisedfrom animals in Fig. 5: (A) liver, (B)spleen, (C) lungs, (D) ovaries, and(E) heart, as determined by qPCR forthe hexon gene, following systemicvector delivery. Error bars, SEM;n ¼ 5/group; ns, P > 0.05; � , P < 0.05;�� , P < 0.01; ��� , P < 0.001;���� , P < 0.0001. Numbers belowgraphs indicate fold decrease ofthe Ad5NULL-A20 group relative tothe Ad5 group.

    Ad5NULL-A20: An Exquisitely Tumor-Selective Virotherapy

    www.aacrjournals.org Clin Cancer Res; 24(17) September 1, 2018 4221

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • Clinical efficacy of therapeutic Ad5 vectors with unmodifiedcapsids is also significantly limited by off-target tissue sequestra-tion, particularly in the liver. We demonstrate that Ad5NULL-A20significantly altered the biodistributionof theAd5 vector in vivobyreducing the sequestration in remarkable magnitudes. In tumor-free mice, replication-deficient Ad5NULL-A20 demonstrated sig-nificantly reduced viral transgene expression the liver, spleen, andlungs compared with the parental Ad5 (Fig. 5), and lower viralgenome copy-number in all off-target organs relative to the Ad5vector (Fig. 6).

    To test the efficacy of an oncolytic form of our detargeted/retargeted Ad5NULL-A20 vector, we established an orthotopic i.p.

    xenograft model of human EOC SKOV3 in immunocompro-misedmice. Themore localized biodistributionof virally encodedtransgene expression of oncolytic Ad5NULL-A20 following intra-peritoneal administration was consistent with reduced off-targetsequestration and/or tumor-selective virus uptake (Fig. 7B–E).This was supported by the superior survival of animals treatedwith Ad5NULL-A20 relative to Ad5 in a SKOV3 xenograft model(Fig. 7E), although extended survival (compared with unmodi-fiedAd5)was not observed inmice treatedwith the oncolytic Ad5.A20 variant. This observation highlights that efficacy in vivodepends upon both the combination of complete ablation of allnative means of cellular uptake via hCAR, avb3/5 integrins and

    Figure 7.

    Oncolytic efficacy study:intraperitoneal delivery of oncolyticvectors in ovarian cancer xenograftmodel. A, Study schedule.Intraperitoneal xenografts of humanovarian cancer SKOV3 cells wereimplanted into immune-compromisedmice (n¼ 5/group), then animalsweretreated with 3 doses of intravenousoncolytic Ad5, avb6 integrinretargeted Ad5.A20 or triplydetargeted, avb6 integrin retargetedAd5NULL-A20, on days 14, 16, and 18. B,Luminescence heatmap images andquantitation of total bodyluminescence were determined at 48hours after the first treatment (C; day16), and at 7 days after the firsttreatment (D; day 21). E, Overallsurvival of animals inoculated withSKOV3 xenografts (avb6-low/CARþ)and then treated with virus, as above,shown as a Kaplan–Meyer survivalcurve until the final study endpoint of101 days. i.p., intraperitoneal; IVIS, invivo imaging system; vp, viral particle;� , P < 0.05; �� , P < 0.01; ��� , P < 0.001.

    Uusi-Kerttula et al.

    Clin Cancer Res; 24(17) September 1, 2018 Clinical Cancer Research4222

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • FX, coupled with an efficient and selective retargeting mechanismto tumor-associated ligands, such as the avb6: A20 receptor:ligand interaction. This observation likely explains previous stud-ies (51, 52), which described no improved efficacy (comparedwith oncolytic Ad5) of virotherapies targeted to avb6 integrin,because the vectors used in those studies lacked modifications inat least two of the three native infectious pathways (the hexon: FXand penton base: avb3/5 interactions). Additional studies will beneeded to fully evaluateavb6þ cancer retargeting in vivo, as well asto dissect the fate in tissues and immunologic responses to theAd5NULL-A20 vector.

    Local, i.p. Ad5NULL-A20 administration presents a promisingtreatment option for advanced, chemotherapy-resistant, avb6þ

    ovarian cancer. Here, we describe a novel vector that can befurther manipulated for various clinical applications, with thescope of selective targeting to avb6 integrin-expressing cellsand minimal off-target effects that limit current Ad5-basedtherapies. Ad5NULL-A20 vector provides an agile and versatileplatform that could ultimately be modified for precisionvirotherapy applications by various innovative approaches,potentially providing a platform for the local, tumor-selectiveoverexpression of additional, virally encoding therapeuticmodalities, such as immunotherapies.

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

    Authors' ContributionsConception and design: R. Jones, R.G. Vile, A.L. ParkerDevelopment of methodology: H. Uusi-Kerttula, J.A. Davies, P. Wongthida,K.G. Shim, A.T. Baker, A.L. ParkerAcquisition of data (provided animals, acquired and managed patients,provided facilities, etc.): H. Uusi-Kerttula, J.A. Davies, J.M. Thompson,

    P. Wongthida, L. Evgin, K.G. Shim, A. Bradshaw, A.T. Baker, R. Jones, L. Hanna,E. Hudson, R.G. VileAnalysis and interpretation of data (e.g., statistical analysis, biostatistics,computational analysis):H.Uusi-Kerttula, J.A.Davies, A. Bradshaw, A.T. Baker,P.J. Rizkallah, R.G. Vile, J.D. Chester, A.L. ParkerWriting, review, and/or revision of the manuscript: H. Uusi-Kerttula,J.A. Davies, P.Wongthida, A.T. Baker, R. Jones, L. Hanna, E. Hudson, J.D. Chester,A.L. ParkerStudy supervision: A.L. Parker

    AcknowledgmentsWe are most grateful to patients at Velindre Cancer Centre, Cardiff, UK, who

    donated ascites samples. We would like to thank Mrs. Dawn Roberts fortechnical assistance, Dr. Richard Stanton for his expertise in AdZ recombineer-ing, Dr. Alexander Greenshields-Watson for his assistance with homologymodeling and structural biology, the team who maintain the YASARA energyminimization server, Dr. EdwardWang for his guidance in immunohistochem-istry, Dr. Lisa Spary and clinicians at Velindre Cancer Centre for access to clinicalascites samples, Mr. William Matchett for his help with the IVIS 200 software,and Prof. Gavin Wilkinson, Dr. Michael Barry, and Dr. John Marshall forinsightful discussions.

    H. Uusi-Kerttula was supported by a Cancer Research Wales PhD stu-dentship to A.L. Parker, J.A. Davies is supported by a Cancer ResearchUK Biotherapeutics Drug Discovery Project Award to A.L. Parker (projectreference C52915/A23946). A.T. Baker is supported by a Tenovus CancerCare PhD studentship to A.L. Parker (project reference PhD2015/L13).A.L. Parker is funded by Higher Education Funding Council for Wales.

    The costs of publication of this article were defrayed in part by thepayment of page charges. This article must therefore be hereby markedadvertisement in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

    Received April 8, 2018; revised May 11, 2018; accepted May 15, 2018;published first May 24, 2018.

    References1. WCRF. Ovarian cancer statistics: World Cancer Research Fund Internation-

    al; 2012[25 June 2016]. Available from: http://www.wcrf.org/int/cancer-facts-figures/data-specific-cancers/ovarian-cancer-statistics.

    2. Yeung TL, Leung CS, Yip KP, Au Yeung CL, Wong ST, Mok SC. Cellular andmolecular processes in ovarian cancermetastasis. a review in the theme: celland molecular processes in cancer metastasis. Am J Physiol Cell Physiol2015;309:C444–56.

    3. Koonings PP, Campbell K, Mishell DR Jr, Grimes DA. Relative frequency ofprimary ovarian neoplasms: a 10-year review. Obstet Gynecol1989;74:921–6.

    4. Koopman Van Aarsen LA, Leone DR, Ho S, Dolinski BM, McCoon PE,LePage DJ, et al. Antibody-mediated blockade of integrin avb 6 inhibitstumor progression in vivo by a transforming growth factor-b-regulatedmechanism. Cancer Res 2008;68:561–70.

    5. Ahmed N, Riley C, Rice GE, Quinn MA, Baker MS. avb6 integrin-A markerfor the malignant potential of epithelial ovarian cancer. J HistochemCytochem 2002;50:1371–9.

    6. Bates RC, Bellovin DI, Brown C, Maynard E, Wu B, Kawakatsu H, et al.Transcriptional activation of integrin b6 during the epithelial-mesenchy-mal transition defines a novel prognostic indicator of aggressive coloncarcinoma. J Clin Invest 2005;115:339–47.

    7. Hazelbag S, Kenter GG, Gorter A, Dreef EJ, Koopman LA, Violette SM,et al. Overexpression of the avb6 integrin in cervical squamouscell carcinoma is a prognostic factor for decreased survival. J Pathol2007;212:316–24.

    8. Cantor DI, CherukuHR, Nice EC, Baker MS. Integrin avb6 sets the stage forcolorectal cancer metastasis. Cancer Metastasis Rev 2015;34:715–34.

    9. Weinacker A, Chen A, AgrezM, Cone RI, Nishimura S, Wayner E, et al. Roleof the integrin avb6 in cell attachment to fibronectin: heterologous

    expression of intact and secreted forms of the receptor. J Biol Chem1994;269:6940–8.

    10. Khan Z, Marshall JF. The role of integrins in TGFbeta activation in thetumour stroma. Cell Tissue Res 2016;365:657–73.

    11. FDA. FDA approves first-of-its-kind product for the treatment ofmelanoma: U.S. Food and Drug Administration; 2015 [Available from:http://www.fda.gov/NewsEvents/Newsroom/PressAnnouncements/ucm469571.htm.

    12. Bourgeois-Daigneault MC, Roy DG, Aitken AS, El Sayes N, Martin NT,Varette O, et al. Neoadjuvant oncolytic virotherapy before surgery sensi-tizes triple-negative breast cancer to immune checkpoint therapy. Sci TranslMed 2018;10:eaao1641.

    13. Samson A, Scott KJ, Taggart D, West EJ, Wilson E, Nuovo GJ, et al.Intravenous delivery of oncolytic reovirus to brain tumor patients immu-nologically primes for subsequent checkpoint blockade. Sci Transl Med2018;10:eaam7577.

    14. clinicaltrials.gov. clinicalTrials.gov: U.S. National Institutes of Health;2016[Available from: www.clinicaltrials.gov.

    15. Bett AJ, Prevec L, Graham FL. Packaging capacity and stability of humanadenovirus type 5 vectors. J Virol 1993;67:5911–21.

    16. Mast TC, Kierstead L, Gupta SB, Nikas AA, Kallas EG, Novitsky V, et al.International epidemiology of humanpre-existing adenovirus (Ad) type-5,type-6, type-26 and type-36 neutralizing antibodies: correlates of high Ad5titers and implications for potential HIV vaccine trials. Vaccine2010;28:950–7.

    17. Parker AL, Waddington SN, Nicol CG, Shayakhmetov DM, Buckley SM,Denby L, et al. Multiple vitamin K-dependent coagulation zymogenspromote adenovirus-mediated gene delivery to hepatocytes. Blood 2006;108:2554–61.

    Ad5NULL-A20: An Exquisitely Tumor-Selective Virotherapy

    www.aacrjournals.org Clin Cancer Res; 24(17) September 1, 2018 4223

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http:&sol;&sol;www.wcrf.org&sol;int&sol;cancer-facts-figures&sol;data-specific-cancers&sol;ovarian-cancer-statisticshttp:&sol;&sol;www.wcrf.org&sol;int&sol;cancer-facts-figures&sol;data-specific-cancers&sol;ovarian-cancer-statisticshttp:&sol;&sol;www.fda.gov&sol;NewsEvents&sol;Newsroom&sol;PressAnnouncements&sol;ucm469571.htmhttp:&sol;&sol;www.fda.gov&sol;NewsEvents&sol;Newsroom&sol;PressAnnouncements&sol;ucm469571.htmwww.clinicaltrials.govhttp://clincancerres.aacrjournals.org/

  • 18. Uusi-Kerttula H, Hulin-Curtis S, Davies J, Parker AL. Oncolytic adenovirus:strategies and insights for vector design and immuno-oncolytic applica-tions. Viruses 2015;7:6009–42.

    19. Waddington SN, McVey JH, Bhella D, Parker AL, Barker K, Atoda H, et al.Adenovirus serotype 5 hexon mediates liver gene transfer. Cell 2008;132:397–409.

    20. Bergelson JM, Cunningham JA, Droguett G, Kurt-Jones EA, Krithivas A,Hong JS, et al. Isolation of a common receptor for Coxsackie B viruses andadenoviruses 2 and 5. Science 1997;275:1320–3.

    21. Coyne CB, Bergelson JM. CAR: a virus receptor within the tight junction.Adv Drug Deliv Rev 2005;57:869–82.

    22. Li Y, Pong RC, Bergelson JM, Hall MC, Sagalowsky AI, Tseng CP, et al.Loss of adenoviral receptor expression in human bladder cancer cells:a potential impact on the efficacy of gene therapy. Cancer Res 1999;59:325–30.

    23. Pearson AS, Koch PE, Atkinson N, Xiong M, Finberg RW, Roth JA, et al.Factors limiting adenovirus-mediated gene transfer into human lung andpancreatic cancer cell lines. Clin Cancer Res 1999;5:4208–13.

    24. Asaoka K, Tada M, Sawamura Y, Ikeda J, Abe H. Dependence of efficientadenoviral gene delivery in malignant glioma cells on the expressionlevels of the Coxsackievirus and adenovirus receptor. J Neurosurg2000;92:1002–8.

    25. Kim JS, Lee SH, Cho YS, Choi JJ, Kim YH, Lee JH. Enhancement of theadenoviral sensitivity of humanovarian cancer cells by transient expressionof coxsackievirus and adenovirus receptor (CAR). Gynecol Oncol2002;85:260–5.

    26. KimM, Zinn KR, Barnett BG, Sumerel LA, Krasnykh V, Curiel DT, et al. Thetherapeutic efficacy of adenoviral vectors for cancer gene therapy is limitedby a low level of primary adenovirus receptors on tumour cells. Eur J Cancer2002;38:1917–26.

    27. DiCara D, Rapisarda C, Sutcliffe JL, Violette SM,Weinreb PH, Hart IR, et al.Structure-function analysis of Arg-Gly-Asp helix motifs in avb6 integrinligands. J Biol Chem 2007;282:9657–65.

    28. Dicara D, Burman A, Clark S, Berryman S, Howard MJ, Hart IR, et al. Foot-and-mouth disease virus forms a highly stable, EDTA-resistant complexwith its principal receptor, integrin alphavbeta6: implications for infec-tiousness. J Virol 2008;82:1537–46.

    29. Stanton RJ, McSharry BP, Armstrong M, Tomasec P, Wilkinson GWG. Re-engineering adenovirus vector systems to enable high-throughput analysesof gene function. BioTechniques 2008;45:659–68.

    30. Uusi-Kerttula H, Davies J, Coughlan L, Hulin-Curtis S, Jones R, Hanna L,et al. Pseudotyped alphavbeta6 integrin-targeted adenovirus vectors forovarian cancer therapies. Oncotarget 2016;7:27926–37.

    31. Uusi-Kerttula H, Legut M, Davies J, Jones R, Hudson E, Hanna L, et al.Incorporation of peptides targeting EGFR and FGFR1 into the adenoviralfiber knob domain and their evaluation as targeted cancer therapies. HumGene Ther 2015;26:320–9.

    32. Fueyo J, Gomez-Manzano C, Alemany R, Lee PSY,McDonnell TJ,MitliangaP, et al. A mutant oncolytic adenovirus targeting the Rb pathway producesanti-glioma effect in vivo. Oncogene 2000;19:2–12.

    33. Gros A, Martínez-Quintanilla J, Puig C, Guedan S, Molleví DG, Alemany R,et al. Bioselection of a gain of functionmutation that enhances adenovirus 5release and improves its antitumoral potency. Cancer Res 2008;68:8928–37.

    34. XiaD, Henry LJ, Gerard RD,Deisenhofer J. Crystal structure of the receptor-binding domain of adenovirus type 5 fiber protein at 1.7 Å resolution.Structure 1994;2:1259–70.

    35. Kotecha A, Wang Q, Dong X, Ilca SL, Ondiviela M, Zihe R, et al. Rulesof engagement between avb6 integrin and foot-and-mouth disease virus.Nat Commun 2017;8:15408.

    36. Emsley P, Lohkamp B, Scott WG, Cowtan K. Features and developmentof Coot. Acta Crystallogr D Biol Crystallogr 2010;66:486–501.

    37. Schr€odinger L. The PyMOL Molecular Graphics System. 2.0 ed.38. Krieger E, Koraimann G, Vriend G. Increasing the precision of comparative

    models with YASARA NOVA—a self-parameterizing force field. Proteins2002;47:393–402.

    39. Krissinel E, Henrick K. Inference of macromolecular assemblies fromcrystalline state. J Mol Biol 2007;372:774–97.

    40. Hynes RO. Integrins: bidirectional, allosteric signaling machines. Cell2002;110:673–87.

    41. Stallwood Y, Fisher KD, Gallimore PH, Mautner V. Neutralisation ofadenovirus infectivity by ascitic fluid from ovarian cancer patients. GeneTher 2000;7:637–43.

    42. Alba R, Bradshaw AC, Parker AL, Bhella D, Waddington SN, Nicklin SA,et al. Identification of coagulation factor (F)X binding sites on the ade-novirus serotype 5 hexon: Effect of mutagenesis on FX interactions andgene transfer. Blood 2009;114:965–71.

    43. Allen MD, Thomas GJ, Clark S, Dawoud MM, Vallath S, Payne SJ, et al.Altered microenvironment promotes progression of preinvasive breastcancer: myoepithelial expression of avb6 integrin in DCIS identifieshigh-risk patients and predicts recurrence. Clin Cancer Res 2014;20:344–57.

    44. Hausner SH, Abbey CK, Bold RJ, Gagnon MK, Marik J, Marshall JF, et al.Targeted in vivo imaging of integrin alphavbeta6 with an improvedradiotracer and its relevance in a pancreatic tumor model. Cancer Res2009;69:5843–50.

    45. Kogelberg H, Tolner B, Thomas GJ, Di Cara D, Minogue S, Ramesh B, et al.Engineering a single-chain Fv antibody to alpha v beta 6 integrin using thespecificity-determining loop of a foot-and-mouth disease virus. J Mol Biol2008;382:385–401.

    46. Whilding LM, Vallath S, Maher J. The integrin alphavbeta6: a noveltarget for CAR T-cell immunotherapy? Biochem Soc Trans 2016;44:349–55.

    47. Moore KM, Thomas GJ, Duffy SW, Warwick J, Gabe R, Chou P, et al.Therapeutic targeting of integrin avb6 in breast cancer. J Natl Cancer Inst2014;106:dju169–dju.

    48. D'Souza SE, Ginsberg MH, Plow EF. Arginyl-glycyl-aspartic acid (RGD): acell adhesion motif. Trends Biochem Sci 1991;16:246–50.

    49. Blackwell JL, Hui L, Gomez-Navarro J, Dmitriev I, Krasnykh V,Richter CA, et al. Using a tropism-modified adenoviral vector tocircumvent inhibitory factors in ascites fluid. Hum Gene Ther 2000;11:1657–69.

    50. Hemminki A, Wang M, Desmond RA, Strong TV, Alvarez RD, Curiel DT.Serum and ascites neutralizing antibodies in ovarian cancer patientstreated with intraperitoneal adenoviral gene therapy. Hum Gene Ther2002;13:1505–14.

    51. Man YKS, Davies JA, Coughlan L, Pantelidou C, Blazquez-Moreno A,Marshall JF, et al. The novel oncolytic adenoviral mutant Ad5–3Delta-A20T retargeted to alphavbeta6 integrins efficiently eliminates pancreaticcancer cells. Mol Cancer Ther 2018;17:575–87.

    52. Coughlan L, Vallath S, Gros A, Gimenez-Alejandre M, Van Rooijen N,Thomas GJ, et al. Combined fiber modifications both to target alpha(v)beta(6) and detarget the coxsackievirus-adenovirus receptorimprove virus toxicity profiles in vivo but fail to improve antitumoralefficacy relative to adenovirus serotype 5. Hum Gene Ther 2012;23:960–79.

    53. Smith T, Idamakanti N, Kylefjord H, Rollence M, King L, Kaloss M, et al. Invivo hepatic adenoviral gene delivery occurs independently of the coxsack-ievirus-adenovirus receptor. Mol Ther 2002;5:770–9.

    Clin Cancer Res; 24(17) September 1, 2018 Clinical Cancer Research4224

    Uusi-Kerttula et al.

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/

  • 2018;24:4215-4224. Published OnlineFirst May 24, 2018.Clin Cancer Res Hanni Uusi-Kerttula, James A. Davies, Jill M. Thompson, et al. Oncolytic Adenovirus for Epithelial Ovarian Cancer Therapies

    6 Integrin-Selectiveβvα-A20: A Tropism-Modified, NULLAd5

    Updated version

    10.1158/1078-0432.CCR-18-1089doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2018/05/24/1078-0432.CCR-18-1089.DC1

    Access the most recent supplemental material at:

    Cited articles

    http://clincancerres.aacrjournals.org/content/24/17/4215.full#ref-list-1

    This article cites 49 articles, 17 of which you can access for free at:

    Citing articles

    http://clincancerres.aacrjournals.org/content/24/17/4215.full#related-urls

    This article has been cited by 2 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://clincancerres.aacrjournals.org/content/24/17/4215To request permission to re-use all or part of this article, use this link

    on August 16, 2020. © 2018 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from

    Published OnlineFirst May 24, 2018; DOI: 10.1158/1078-0432.CCR-18-1089

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-18-1089http://clincancerres.aacrjournals.org/content/suppl/2018/05/24/1078-0432.CCR-18-1089.DC1http://clincancerres.aacrjournals.org/content/24/17/4215.full#ref-list-1http://clincancerres.aacrjournals.org/content/24/17/4215.full#related-urlshttp://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/24/17/4215http://clincancerres.aacrjournals.org/

    /ColorImageDict > /JPEG2000ColorACSImageDict > /JPEG2000ColorImageDict > /AntiAliasGrayImages false /CropGrayImages false /GrayImageMinResolution 200 /GrayImageMinResolutionPolicy /Warning /DownsampleGrayImages true /GrayImageDownsampleType /Bicubic /GrayImageResolution 300 /GrayImageDepth -1 /GrayImageMinDownsampleDepth 2 /GrayImageDownsampleThreshold 1.50000 /EncodeGrayImages true /GrayImageFilter /DCTEncode /AutoFilterGrayImages true /GrayImageAutoFilterStrategy /JPEG /GrayACSImageDict > /GrayImageDict > /JPEG2000GrayACSImageDict > /JPEG2000GrayImageDict > /AntiAliasMonoImages false /CropMonoImages false /MonoImageMinResolution 600 /MonoImageMinResolutionPolicy /Warning /DownsampleMonoImages true /MonoImageDownsampleType /Bicubic /MonoImageResolution 900 /MonoImageDepth -1 /MonoImageDownsampleThreshold 1.50000 /EncodeMonoImages true /MonoImageFilter /CCITTFaxEncode /MonoImageDict > /AllowPSXObjects false /CheckCompliance [ /None ] /PDFX1aCheck false /PDFX3Check false /PDFXCompliantPDFOnly false /PDFXNoTrimBoxError true /PDFXTrimBoxToMediaBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXSetBleedBoxToMediaBox true /PDFXBleedBoxToTrimBoxOffset [ 0.00000 0.00000 0.00000 0.00000 ] /PDFXOutputIntentProfile (None) /PDFXOutputConditionIdentifier () /PDFXOutputCondition () /PDFXRegistryName () /PDFXTrapped /False

    /CreateJDFFile false /Description > /Namespace [ (Adobe) (Common) (1.0) ] /OtherNamespaces [ > /FormElements false /GenerateStructure false /IncludeBookmarks false /IncludeHyperlinks false /IncludeInteractive false /IncludeLayers false /IncludeProfiles false /MarksOffset 18 /MarksWeight 0.250000 /MultimediaHandling /UseObjectSettings /Namespace [ (Adobe) (CreativeSuite) (2.0) ] /PDFXOutputIntentProfileSelector /NA /PageMarksFile /RomanDefault /PreserveEditing true /UntaggedCMYKHandling /LeaveUntagged /UntaggedRGBHandling /LeaveUntagged /UseDocumentBleed false >> > ]>> setdistillerparams> setpagedevice