throbber
Downloaded from http://aacrjournals.org/clincancerres/article-pdf/15/8/2777/1988959/2777.pdf by guest on 26 September 2022
`
`CancerTherapy: Preclinical
`
`Efficacy of Systemically Administered Oncolytic Vaccinia
`Virotherapy for Malignant Gliomas Is Enhanced by Combination
`Therapy with Rapamycin or Cyclophosphamide
`Xue Qing Lun,1,2 Ji-Hyun Jang,3 Nan Tang,3 Helen Deng,3 Renee Head,3 John C. Bell,6 David F. Stojdl,7
`Catherine L. Nutt,8,9 Donna L. Senger,1,2 Peter A. Forsyth,1,2 and J. Andrea McCart3,4,5
`
`Abstract Purpose: The oncolytic effects of a systemically delivered, replicating, double-deleted vaccinia
`virus has been previously shown for the treatment of many cancers, including colon, ovarian,
`and others.The purpose of this study was to investigate the oncolytic potential of double-deleted
`vaccinia virus alone or in combination with rapamycin or cyclophosphamide to treat malignant
`gliomas in vitro and in vivo.
`Experimental Design: Rat (RG2, F98, C6) and human (A172, U87MG, U118) glioma cell
`lines were cultured in vitro and treated with live or UV-inactivated vaccinia virus. Viral gene
`[enhanced green fluorescent protein (EGFP)] expression by fluorescence-activated cell
`sorting, relative cell viability by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide
`(MTT), and assays for cytopathic effects were examined. S.c. murine tumor xenografts
`(U87MG, U118, C6) and i.c. (RG2, F98) tumor models in immunocompetent rats were treated
`with systemic administration of EGFP-expressing vaccinia virus (vvDD-EGFP), alone or in
`combination with rapamycin or cyclophosphamide, or controls. Tumor size, viral biodistribution,
`and animal survival were assessed. Lastly, the oncolytic effects of vvDD-EGFP on human malig-
`nant glioma explants were evaluated.
`Results: vvDD-EGFP was able to infect and kill glioma cells in vitro. A single systemic dose
`of vvDD-EGFP significantly inhibited the growth of xenografts in athymic mice. Systemic delivery
`of vvDD-EGFP alone was able to target solitary and multifocal i.c. tumors and prolong survival of
`immunocompetent rats, whereas combination therapy with rapamycin or cyclophosphamide
`enhanced viral replication and further prolonged survival. Finally, vvDD-EGFP was able to infect
`and kill ex vivo primary human malignant gliomas.
`Conclusions: These results suggest that vvDD-EGFP is a promising novel agent for human
`malignant glioma therapy, and in combination with immunosuppressive agents, may lead to
`prolonged survival from this disease.
`
`There has been minimal
`in the median
`improvement
`survival of patients with malignant gliomas, and these
`tumors remain largely incurable despite advances in surgery,
`radiation, and chemotherapy (1). The need for new
`treatment strategies for malignant gliomas has led to the
`emergence of oncolytic virotherapy, oncolytic viruses able
`to selectively destroy tumor cells while sparing normal
`tissue. Much effort to date has been achieved in preclinical
`
`models of malignant gliomas using several oncolytic viruses,
`including herpes virus (2), adenovirus (3),
`reovirus (4),
`poliovirus (5), myxoma virus (6), and vesicular stomatitis
`virus (7). Some of these have already been tested in early
`clinical trials (8 – 10), and a small number of responses were
`found. Efficacy may be limited by factors such as the host
`immune response against the virus (11, 12) and limitations
`of systemic delivery in brain tumor patients, which may
`
`Authors’ Affiliations: 1Departments of Oncology, Clinical Neurosciences,
`Biochemistry and Molecular Biology, Tom Baker Cancer Centre; 2Clark H. Smith
`Brain Tumor Center, University of Calgary, Alberta, Canada; 3Division of
`Experimental Therapeutics, Toronto General Research Institute; 4Department of
`Surgery, Mount Sinai Hospital; 5University of Toronto, Toronto, Ontario, Canada;
`6Centre for Cancer Therapeutics, Ottawa Health Research Institute; 7Apoptosis
`Research Centre, Children’s Hospital of Eastern Ontario, Ottawa, Ontario, Canada;
`8Department of Pathology, Massachusetts General Hospital; and 9Harvard Medical
`School, Boston, Massachusetts
`Received 9/9/08; revised12/8/08; accepted12/9/08; published OnlineFirst 4/7/09.
`Grant support: National Cancer Institute of Canada with funds raised by the
`Canadian Cancer Society (P.A. Forsyth and D.L. Senger); a Program Project Grant
`from theTerry Fox Foundation (P.A. Forsyth, D.L. Senger, J.C. Bell, and D.F. Stojdl);
`the Clark Smith BrainTumor Center (P.A. Forsyth); a Grant Miller Cancer Research
`
`Grant from the Faculty of Medicine, University of Toronto (J.A. McCart); and
`commercial support from Jennerex (D.F. Stojdl and J.C. Bell).
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance
`with 18 U.S.C. Section 1734 solely to indicate this fact.
`Note: Supplementary data for this article are available at Clinical Cancer Research
`Online (http://clincancerres.aacrjournals.org/).
`P.A. Forsyth and J.A. McCart share senior authorship.
`Requests for reprints: J. Andrea McCart, Division of Experimental Therapeutics,
`Toronto General Research Institute, Room 4-408, 67 College Street, Toronto,
`Ontario, Canada. Phone: 416-586-4552; Fax: 416-586-8392; E-mail: amccart@
`uhnres.utoronto.ca.
`F 2009 American Association for Cancer Research.
`doi:10.1158/1078-0432.CCR-08-2342
`
`www.aacrjournals.org
`
`2777
`
`Clin Cancer Res 2009;15(8) April 15, 2009
`
`Replimune Limited Ex. 2022 - Page 1
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/15/8/2777/1988959/2777.pdf by guest on 26 September 2022
`
`were to determine the following: (a) the efficacy of an EGFP-
`expressing vaccinia virus (vvDD-EGFP) alone or in combina-
`tion with rapamycin or cyclophosphamide as an experimental
`therapeutic agent against malignant gliomas in vitro and in vivo,
`and (b) the distribution and clearance of the virus after systemic
`(i.v.) delivery in immunocompetent hosts. We found that
`vvDD-EGFP targeted malignant glioma tumors specifically and
`prolonged survival in immunocompetent animal models of
`malignant glioma. This was further improved by the addition of
`rapamycin and cyclophosphamide. Although no cures were
`seen, there was a dramatic improvement in median survival. We
`also found that systemic (i.v.) administration of vvDD-EGFP in
`rats was feasible, safe, and well tolerated. The results of this
`study suggest
`that
`further investigation of
`the utility of
`oncolytic viruses in the treatment of malignant gliomas is
`warranted.
`
`Materials and Methods
`
`Cell lines. Human and rat glioma cell lines (U87MG, U118MG,
`A172, F98, RG2, and C6), monkey kidney fibroblasts (CV1), and
`murine NIH3T3 cells were obtained from the American Type Culture
`Collection. All cell lines were grown in DMEM supplemented with 10%
`heat-inactivated FCS, 2 mmol/L glutamine, and 1% antibiotic-
`antimycotic (Gibco). Cell lines were maintained in an incubator at
`37jC with 5% CO2 and serially passaged every 3 to 4 d. Each cell line
`was tested routinely for mycoplasma contamination.
`Vaccinia viruses. The recombinant (WR strain) vaccinia viruses
`vvDD-EGFP (14) and vvDD-R2RLuc were used in these studies. vvDD-
`R2RLuc was constructed from the plasmid pTREX-R2RLuc, which
`contains the red fluorescent protein (mCherry; gene synthesized
`commercially) fused through a foot-and-mouth disease virus 2A
`peptide motif to the Renilla luciferase gene (Promega Corp.). The
`foot-and-mouth disease virus 2A motif allows for bicistronic expression
`of the two genes. The R2RLuc segment was subcloned into our vaccinia
`shuttle plasmid pVx – EGFP (14) upstream of the vaccinia synthetic
`early/late promoter, replacing EGFP. After homologous recombination
`with the parental virus VSC20 (ref. 29; a gift from Dr. B. Moss), vvDD-
`R2RLuc underwent five rounds of selection in mycophenolic acid. Both
`viruses are based on the double-deleted platform, which lacks
`thymidine kinase and vaccinia growth factor described previously
`(14). All viruses were expanded in HeLa cells, purified on a sucrose
`cushion, and tittered on CV1 cells (30).
`Flow cytometry analysis. Cells were infected with vvDD-EGFP
`or mock control at a multiplicity of
`infection (MOI) of 1 and
`harvested after 0, 24, 48, and 72 h. Cells were washed twice with
`
`1 PBS, resuspended in fluorescence-activated cell sortings (FACS)
`
`buffer (Optimized Sheath Fluid, BD Biosciences), and evaluated by
`FACS caliber (Becton Dickinson Canada, Inc.). Green fluorescent
`protein (FL1; 488 nm) and phycoerythrin (PE) (FL2; 488 nm;
`compensation, FL1-1.1% FL2) expression was analyzed using Cell-
`Quest software (BD Biosciences).
`Cell proliferation assays. For the MTT assay, cells were infected with
`vvDD-EGFP at an MOI of 1, seeded in complete medium (10% FCS)
`
`into 96-well plates (5  105 cells/well; triplicate), and incubated at
`37jC in a 5% CO2 humidified atmosphere. After 0, 24, 48, and 72 h,
`10 AL of the MTT labeling reagent (5 mg/mL in PBS; Cell Proliferation
`Kit 1, Roche Applied Science) was added to each well. After 4 h (37jC;
`5% CO2), 100 AL of solubilization solution (10% SDS in 0.01 mol/L
`HCL) was added and cells were incubated overnight. Microtiter plates
`were evaluated on a Vmax Kinetic microplate reader (Molecular Devices
`Corp.), and data analysis was done (SOFTMax software version 2.32,
`Molecular Devices Corp.). For the combined therapy group, rapamycin
`
`CancerTherapy: Preclinical
`
`Translational Relevance
`
`Malignant gliomas, an aggressive form of brain tumor,
`are currently incurable. In this study, we examine the activity
`of a novel therapeutic double-deleted vaccinia virus
`(vvDD). We show in animal models that vvDD is effective
`in prolonging survival from malignant gliomas and it is
`enhanced by the chemotherapy agents cyclophosphamide
`and rapamycin, which are now in clinical use. vvDD is
`safe in these models and is an excellent candidate to take
`forward in clinical trials for malignant glioma. This is
`an important step toward the future development of
`new treatments for malignant gliomas using this type of
`combination therapy.
`
`include the blood-brain barrier (13) and immune responses
`to virus in the vascular compartment.
`Vaccinia virus is a double-stranded, enveloped, lytic DNA
`virus (14) with several advantages over other oncolytic viruses.
`It is easy to manipulate genetically; its replication and spread
`are rapid; and it is motile (actin tail dependent) and has no
`potential capacity to integrate into foreign DNA. It is a clinically
`safe and well-known virus because of its widespread use as a
`vaccine in the small pox eradication program. Several poxvirus-
`based vaccination trials for cancer are currently under
`investigation (15, 16), highlighting the relevance of this virus
`for anticancer immunotherapy.
`Many strains of attenuated replicating vaccinia viruses have
`shown promising efficacy in the treatment of murine models of
`human gliomas (17) and other cancers (14, 18, 19), as well as
`primary cultures of human tumors (20). Because there may be a
`potential concern about the safety of a replicating vaccinia
`virus, a mutant ‘‘double-deleted’’ version of the Western Reserve
`(WR) strain [double-deleted vaccinia virus (vvDD)] with
`deletions of the thymidine kinase and vaccinia growth factor
`genes was created to enhance its safety (14). vvDD was
`nontoxic following i.v. delivery in nonhuman primates (21),
`which suggests that it may be a good candidate for the systemic
`oncolytic virotherapy of human tumors.
`As the efficacy of oncolytic virotherapy as a single agent has
`thus far been unsatisfactory (8 – 10), this suggests that new or
`multiple oncolytic viruses or other combination therapies might
`prove to be more effective against malignant gliomas. Others
`have investigated oncolytic viruses in combination with chemo-
`therapeutics, radiation therapy, or with suicide gene/prodrug
`systems, and have found enhanced efficacy of the oncolytic
`viruses toward brain tumors (22 – 24). Improved results have also
`been seen in immunocompetent animals using an oncolytic
`herpes virus in combination with the cyclophosphamide to
`suppress the immune response and provide for prolonged
`replication of the virus (25, 26). Recently, we and others have
`shown that the combination of myxoma virus plus rapamycin led
`to enhanced viral replication in vitro and in vivo, and improved
`efficacy against medulloblastomas (27) and melanoma (28). This
`led us to investigate the use of oncolytic vaccinia virus in
`combination with rapamycin or cyclophosphamide therapy for
`the treatment of experimental models of malignant gliomas.
`To date, vvDD has not been evaluated for its efficacy or
`toxicity in brain tumor models. The objectives of this study
`
`Clin Cancer Res 2009;15(8) April 15, 2009
`
`2778
`
`www.aacrjournals.org
`
`Replimune Limited Ex. 2022 - Page 2
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/15/8/2777/1988959/2777.pdf by guest on 26 September 2022
`
`Vaccinia Therapy of Malignant Glioma
`
`(1-10 nmol/L/mL) was added to the cells 1 h before treatment
`with virus.
`In vitro cytopathic effects (cytopathic effect assay) were visualized in
`six-well plates. Malignant glioma cell lines were plated until confluent
`and then infected with vvDD-EGFP (or mock control or UV-inactivated
`control) at an MOI of 1 for 2 h. At 24, 48, and 72 h after infection,
`cells were photographed under white light (original magnification,
`
`10; Nikon Eclipse TE200 microscope with a Hamamatsu ORCA100
`
`digital camera).
`Animals. CD-1 nude mice (female; 6-8 wks old) and Fisher 344 rats
`were purchased from Charles River Canada. The animals were housed
`in groups of three to five in a vivarium maintained on a 12-h light/dark
`schedule with a temperature of 22jC F 1jC and a relative humidity of
`50% F 5%. Food and water were available ad libitum. All procedures
`were reviewed and approved by the University of Calgary and
`University Health Network (Toronto, Canada) Animal Resource
`Centres.
`In vivo oncolysis in a s.c. xenograft model. To determine the
`antitumor effects of vaccinia virus in s.c. models, we injected C6, U87,
`and U118 (1  107 cells/mouse) malignant glioma cells into the right
`
`flank of female nude mice to establish s.c. tumors. Seven days later,
`when tumor volumes reached 0.10 cm3 for C6, 0.13 cm3 for U87, and
`0.06 cm3 for U118, the treatment group was injected i.p. with 109
`particle-forming unit of vvDD-EGFP in 2 mL of HBSS and the control
`group with 2 mL of HBSS i.p. Tumors were then measured twice a week
`by a blinded investigator, and tumor volume was calculated as (width)2
` length  0.52. Animals were sacrificed according to Animal Resource
`
`Centres guidelines, and tumor tissues were removed for histologic
`examination.
`Evaluation of toxicity of vvDD-EGFP in immunocompetent rats. To
`investigate the toxicity of vvDD-EGFP in non-tumor-bearing immuno-
`competent rats, Fischer 344 rats (n = 12) under anesthesia (isofluorane
`
`2%) received i.c. injections of increasing doses of vvDD-EGFP (1  102,
`1  104, 1  106, 1  108 particle-forming unit/rat; two rats per dose)
`using the guide-screw system (31) or 1  109 particle-forming unit/rat
`
`or vehicle control via tail vein. Animals were followed for 42 d and
`weighed twice per week. Animals losing 20% body weight or having
`other unacceptable symptoms were sacrificed as per our Institutional
`Animal Care Guidelines. After sacrifice, brains and major organs were
`taken out and processed for histologic examination.
`
`Fig. 1. vvDD-EGFP infects human and rat brain tumor cells in vitro. A, fluorescence-activated cell sorting analysis for EGFP of malignant glioma cell lines (C6, A172, RG2,
`U87MG, and F98) 24 to 72 h after infection with the vvDD-EGFP at an MOI of 1. B, MTTassay of malignant glioma cells (human: U87MG, U118MG, A172; rat: C6, RG2, F98)
`compared with uninfected controls over 72 h. C, cytopathic effect on malignant gliomas cells. Cells plated at confluence were infected the next day with vvDD-EGFP or
`UV/psoralen-inactivated virus (UV vvDD) at an MOI of 1. Microscopy was done 48 h after viral infection. Original magnification, 400.
`
`www.aacrjournals.org
`
`2779
`
`Clin Cancer Res 2009;15(8) April 15, 2009
`
`Replimune Limited Ex. 2022 - Page 3
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/15/8/2777/1988959/2777.pdf by guest on 26 September 2022
`
`Survival studies of orthotopic glioma models in immunocompetent
`hosts. To investigate the efficacy of vvDD-EGFP in orthotopic glioma
`animal models, female Fischer 344 rats under anesthesia were injected
`
`with 1  105 F98 cells in 3 AL of sterile PBS using the guide-screw
`system (31) or were fixed to a stereotactic apparatus, and 5  104
`RG2 cells in 3 AL of PBS were inoculated as described previously (6).
`Seven (F98) or five (RG2) days after tumor implantation, rats were
`i.v. administrated vvDD-EGFP (1  109 particle-forming unit/rat in
`100 AL HBSS) as a single dose or multiple doses (1  109 particle-
`
`forming unit/rat every 2 d for a total of three injections). Control
`animals were treated with HBSS. Rats were monitored daily until they
`lost >20% of body weight or had trouble ambulating, feeding, or
`grooming; then, they were sacrificed, and their brains and major organs
`were collected for histologic analysis.
`In vivo viral distribution studies in an orthotopic glioma model. To
`determine if vvDD-EGFP targets multifocal gliomas in the brain, we
`established a bilateral tumor model using RG2 tumor cells to mimic
`multifocal malignant glioma of patients. RG2 cells were implanted by
`stereotactic techniques as described above in immunocompetent Fischer
`rats. Ten days later, rats were administered i.v. vvDD-EGFP
`
`(1  109 particle-forming unit/rat in 100 AL HBSS). Animals (n = 3)
`
`were sacrificed at 1, 3, 5, 7, and 14 d after virus administration. When
`sacrificed, animals were anesthetized and perfused with 50 mL of saline,
`followed by 30 mL of phosphate-buffered 10% formalin via cardiac
`catheter. Following fixation, a whole brain picture was taken with a Leica
`MZ-FLIII fluorescence stereomicroscope using a standard green fluores-
`cent protein filter set and the brain, tumor, and other major organs were
`removed and frozen in liquid nitrogen for virus recovery assays.
`Combination therapy in vitro and in vivo. To determine whether
`pretreatment with rapamycin promotes vvDD oncolysis of rat cell
`lines in vitro, viral green fluorescent protein expression and cell viability
`were assessed 48 or 72 h postinfection in the presence or absence
`of rapamycin. F98 and RG2 Cells were pretreated with rapamycin
`(1 nmol/l) 1 h before virus infection, then infected with vvDD-EGFP
`(MOI, 0.1 or 1). After a 48-h incubation, green fluorescent protein
`expression was analyzed using a Zeiss inverted microscope (Axiovert
`200 M) with green fluorescent protein filter and a Carl Zeiss camera
`(AxioCam MRc). To assess the effects of combined therapy on cell
`viability, an MTT assay was done 72 h after infection. To determine the
`effect of rapamycin on viral replication, cells were pretreated for 2 h with
`rapamycin (10 nmol/L),
`infected with vvDD-EGFP (MOI, 0.01),
`incubated for 48 h, and lysed using three rounds of freeze thawing to
`extract viral particles. The viral titers of samples were determined using a
`standard plaque titration assay on CV1 cells.
`To evaluate the in vivo effects of combination therapy using vvDD-
`EGFP combined with rapamycin or cyclophosphamide, i.c. bilateral
`RG2 tumor – bearing animals (described above) were divided into
`the following four groups (n = 9-10 per group) 8 d after implantation:
`(a) dead virus control, (b) drug alone (i.p. administration of rapamycin
`5 mg/kg/d for 5 d or cyclophosphamide 60 mg/kg, one time only),
`
`(c) vvDD-EGFP (1  109 particle-forming unit/rat) alone, and (d)
`
`vvDD-EGFP plus drug (as above beginning 1 d before virus injection).
`Two animals (of nine) were sacrificed on day 7 to assess green
`fluorescent protein expression. Five or six animals (of 9 or 10) in each
`group were monitored daily and sacrificed when symptoms developed
`as described above.
`To assess the effects of rapamycin and cyclophosphamide on the
`immune response to viral replication, two animals (of nine) from each
`group were pretreated with drug and then treated with a second dose of
`vaccinia 3 d after the initial dose. The second virus expresses red
`
`fluorescent protein (vvDD-RFPLuc; 1  109 particle-forming unit/rat)
`
`to differentiate it from the initial injection. These mice were sacrificed
`3 d after the second virus was administered. The brains were removed
`and photographed using a fluorescent stereotactic microscope (Leica
`MZ-FLIII, Switzerland, Ltd.) to visualize green fluorescent protein – and
`red fluorescent protein – expressing virus, then frozen in liquid nitrogen
`for virus recovery assays.
`
`CancerTherapy: Preclinical
`
`Fig. 2. Systemic (i.p.) administration of vvDD-EGFP on growth of s.c. models of
`malignant glioma in nude mice. Female nude mice were injected with 1 107 C6,
`U87, and U118 cells s.c. in their right flanks. Seven days later, when tumor volumes
`reached f0.10 cm3 for C6, 0.13 cm3 for U87, and 0.06 cm3 for U118, a blinded
`experimenter administered the treatment group with 109 particle-forming units of
`vvDD-EGFP in 2 mL of HBSS i.p. and the control group with 2 mL of HBSS i.p.
`Tumors were then measured twice a week. A, C6 malignant gliomas in the treatment
`group had significantly (P = 0.03) smaller tumor volumes than the control
`group. B, the volume of U87 gliomas in the treatment group remained stable,
`whereas the tumor volume of the control group increased significantly (P = 0.01)
`with time. C, the volume of U118 malignant glioma also remained relatively
`stable over time in the treatment group, whereas the tumor volume of the control
`group increased significantly (P = 0.03).
`
`Clin Cancer Res 2009;15(8) April 15, 2009
`
`2780
`
`www.aacrjournals.org
`
`Replimune Limited Ex. 2022 - Page 4
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/15/8/2777/1988959/2777.pdf by guest on 26 September 2022
`
`Vaccinia Therapy of Malignant Glioma
`
`Immunohistochemistry. Paraffin embedded sections of rat brain
`
`were deparaffinized and rehydrated with 1 PBS after blocking, then
`
`exposed to primary antibody (murine monoclonal vaccinia virus
`antibody, Abcam, Inc.) at a 1:10 dilution in PBS overnight at 4jC.
`Biotinylated donkey anti – mouse IgG (1:2,000, Vector Laboratories)
`was used as the secondary antibody. Sections were then incubated with
`avidin conjugated to horseradish peroxidase (Vectastain ABC immu-
`nohistochemistry kit, Vector Laboratories), and staining was visualized
`by the addition of 3,3¶-diaminobenzidine substrate with hematoxylin
`counterstaining. Sections were mounted and viewed with a Zeiss
`inverted microscope (Axiovert 200M) and a Carl Zeiss camera
`(AxioCam MRc) to obtain images.
`For the CD68/CD163 staining, frozen sections of the brains were
`fixed with 4% paraformaldehyde for 15 to 20 mins, followed by two
`washes with PBS (for paraffin embedded sections were deparaffinized
`
`and rehydrated with 1 PBS). The sections were incubated with
`
`primary antibody [mouse anti – rat CD68 (1:500; Serotec; Cat MCA
`341R), CD163 (1:300; Serotec; Cat MCA 342R)] for 1 h at room
`temperature after blocking. Biotinylated anti – mouse IgG (1:500; Vector
`Laboratories; CatBA-2000) was used as a secondary antibody.
`
`Virus recovery assays. Rats were sacrificed; saline was immediately
`infused; and the tissues were extracted and homogenized in HBSS using
`a Pellet Pestles Kit (VWR International), followed by repeated freeze
`thawing to release virus from the cells. Supernatants were plaque
`tittered on CV1 cells, as previously described (29). Viral plaques were
`counted, and particle-forming unit were calculated by the number of
`plaques multiplied by the dilution factor and normalized to the weight
`of the tissues.
`Primary human glioma culture. Short-term cultures were established
`from patient samples of human gliomas (n = 8) obtained following
`brain tumor surgery at
`the Foothills Hospital (Calgary, Alberta,
`Canada). This study was approved by the Conjoint Medical Ethics
`Committee. Briefly, each patient specimen was split into two pieces for
`fixation in 10% formalin and short-term culture. For short-term
`cultures, the tissue was washed several times in sterile saline, cut into
`small pieces (f0.5-1 mm in diameter), and dissociated with trypsin
`(0.25%) and 50 Ag/mL DNase (Roche Diagnostics) for 30 mins at
`37jC. After filtering and washing with DMEM/F12 (containing 20%
`FBS), cells were resuspended in 20% FBS in DMEM/F12 and plated
`(at 10,000-100,000 cells per well) in 96-well plates. Cells were infected
`
`Fig. 3. Systemic (i.v.) administration of vvDD-EGFP prolonged survival of immunocompetent rats bearing i.c. F98 and RG2 gliomas. A, F98 tumor model. Kaplan-Meier
`survival analysis of rats implanted with F98 (1 105 cells/rat) and treated with either HBSS (n = 6) or vvDD-EGFP (n = 6; 1 109 particle-forming units/rat single i.v. injection
`on day 7 after tumor implantation). All Ps are two sided. Arrow, virus injections. B, representative immunohistochemistry of vvDD analysis in F98 tumors and normal brain
`7 d after i.v. administration of virus. Left column, mock control. Original magnification, 10 (top) and 40 (bottom). C, RG2 tumor model. Kaplan-Meier plot showing
`survival curves of rats harboring i.c. RG2 tumor treated with dead virus (n = 5) or a single i.v. administration of vvDD-EGFP (live virus single; n = 5; 1 109 particle-forming
`units/rat) or multiple i.v. administration of vvDD-EGFP (live virus multiple; n = 6; 1 109/rat every 2 d for three injections). All Ps are two sided. Arrows, day of virus
`administration. D, histologic analysis showed that all of the dead virus ^ treated mice and live virus ^ treated mice had large tumors in the brain. Most of animals treated with
`i.v. administration of vvDD-EGFP showed area of tumor necrosis. Original magnification, 25 (top) and 400 (bottom).
`
`www.aacrjournals.org
`
`2781
`
`Clin Cancer Res 2009;15(8) April 15, 2009
`
`Replimune Limited Ex. 2022 - Page 5
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/15/8/2777/1988959/2777.pdf by guest on 26 September 2022
`
`method. The log-rank test and ANOVA for repeated measures were used
`to compare the effect of different forms of treatment. The Student’s t test
`was used when appropriate. Data was expressed as means F SD. All Ps
`<0.05 were considered significant.
`
`Results
`
`Vaccinia virus (vvDD-EGFP) infects and kills malignant
`glioma in vitro. Malignant glioma cell lines were tested for
`susceptibility to infection by vvDD-EGFP using FACS analysis
`to quantitate the percentage of EGFP-positive cells. Malignant
`glioma cell lines (C6, A172, RG2, U87MG, and F98) were
`infected with vvDD-EGFP at an MOI of 1; the number of
`EGFP-positive cells was quantified 24, 48, and 72 hours after
`infection. All 5 malignant glioma cell lines were efficiently
`infected by vvDD-EGFP, with 70% to 99% of all cell lines
`infected by 24 hours (Fig. 1A).
`To confirm whether infection was leading to oncolysis and
`cell death, MTT and cytopathic effect assays were done. All six
`cell
`lines (U87, U118, A172, C6, RG2, and F98) were
`susceptible to killing, and extensive cell death was observed
`72 h after infection with an MOI of 1 (<40% of cells still viable
`compared with uninfected controls; Fig. 1B). Similar results
`were obtained with the cytopathic effect assay. There was no
`evidence of cytopathic effect in untreated (control) or dead
`virus – treated (UV vvDD) cells at an MOI of 1 at 48 hours
`postinfection (Fig. 1C).
`Antitumor effect of systemic administration of vvDD-EGFP on
`s.c. models of malignant glioma. We next investigated if vvDD-
`EGFP would infect and kill malignant glioma in a s.c. tumor
`model in nude mice. Nude mice bearing s.c. xenografts of
`U87MG, U118, or C6 malignant glioma cell lines were injected
`systemically (i.p.) with 109 particle-forming unit of vvDD-EGFP
`or HBSS control. We observed a statistically significant
`inhibition of tumor growth in virus-treated mice compared
`with the HBSS-treated control mice bearing either C6 (ANOVA;
`P = 0.03; Fig. 2A) or U87 (ANOVA; P = 0.01) tumors (Fig. 2B).
`The volume of U118 malignant glioma remained relatively
`stable over time in the treatment group, whereas the tumor
`volume of the control group increased significantly (Fig. 2C;
`ANOVA; P = 0.03).
`Toxicity of intracerebral and i.v. administration of vvDD-
`EGFP. We evaluated the toxicity of vvDD-EGFP in non-tumor-
`bearing immunocompetent rats when administered i.c. or i.v. A
`
`single dose of 1  108 particle-forming unit i.c. or 1  109
`
`particle-forming unit i.v. (the highest feasible dose we can
`prepare) of vvDD-EGFP was safe in normal non-tumor-bearing
`rats. At the highest i.c. dose administered, we saw some weight
`loss, which quickly recovered (Supplementary Fig. 1). There
`were no deaths and no neurologic symptoms noted. Surpris-
`ingly, when tumor-bearing rats were subsequently treated, all
`i.c. doses were extremely toxic in a non-dose-dependent
`manner. Necropsy showed gross hemorrhage and swelling of
`the tumor, presumably because of the rapid oncolysis by the
`
`virus (data not shown). I.v. administration of 1  109 particle-
`
`forming units to tumor-bearing rats was nontoxic and was the
`dose/route used in subsequent studies.
`Survival following systemic administration of vvDD-EGFP in
`immunocompetent rats bearing i.c. malignant glioma. To
`determine the efficacy of vvDD-EGFP oncolysis in immuno-
`competent models of i.c. malignant glioma, we implanted F98
`
`CancerTherapy: Preclinical
`
`Fig. 4. Distribution of i.v. administered vvDD-EGFP in immunocompetent rats
`bearing bilateral i.c. malignant gliomas. Rats bearing bilateral RG2 tumors (to mimic
`multifocal malignant glioma clinically) were treated with vvDD-EGFP i.v. at a dose
`of 1 109 particle-forming units rat 10 d after implantation of RG2 tumors. Animals
`were sacrificed at different time points (1, 3, 5, 7 d) and 14 d (data not shown)
`after viral administration. A, representative photomicrographs of EGFP ^ labeled
`virus in the brain tumor (n = 2 rat/group). Original magnification, 10 (top) and
`20 (bottom). Arrows, green fluorescent protein virus expression. B, titer of virus
`present in the bilateral tumors (n = 3 rats per time point).Tumors on each side of the
`brain (right or left) or resections of normal brain (brain) were harvested for virus
`extraction, and the samples were analyzed by virus recovery assay on CV1cells.
`C, titers of virus present in other organs in vivo following i.v. administration of
`vvDD-EGFP.
`
`the following day with vvDD-EGFP virus, live and UV inactivated, at
`MOIs of 0.1, 1, and 10. Cell viability was measured 96 h postinfection
`by MTT assay. Viral
`titers were obtained from patient short-term
`cultures after infection. Dissociated tumor cells from surgical samples of
`malignant gliomas were plated in six-well plates and infected the next
`day with vvDD-EGFP at an MOI of 0.1. Cells were collected at 0, 24,
`and 72 h after virus infection. These were then lysed using three rounds
`of freeze thawing to extract viral particles. U87 and NIH3T3 were used
`as positive and negative controls, respectively. The viral titers of samples
`were determined using a standard plaque titration assay on CV1 cells.
`Statistics. All statistics were generated using StatView Software
`(Abacus Concepts, Inc.) and GraphPad Prism (version 4; GraphPad
`Software, Inc.). Survival curves were generated by the Kaplan-Meier
`
`Clin Cancer Res 2009;15(8) April 15, 2009
`
`2782
`
`www.aacrjournals.org
`
`Replimune Limited Ex. 2022 - Page 6
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Vaccinia Therapy of Malignant Glioma
`
`orRG2 cells into the brain of F344 rats to establish i.c. tumors.
`Rats were treated i.v. with single or multiple doses of vvDD-
`EGFP after tumor implantation. Animals were monitored daily.
`A single i.v. administration of vvDD-EGFP prolonged survival
`(Fig. 3A; long-rank test; P = 0.0475) of rats bearing i.c. F98
`tumors. The median survival of vvDD-EGFP – treated animals
`was 24.5 days [95% confidence interval (95% CI), 20-28 days],
`whereas contr

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket