throbber
Cancer Therapy: Preclinical
`
`Defining Effective Combinations of Immune
`Checkpoint Blockade and Oncolytic Virotherapy
`Juan J. Rojas1, Padma Sampath1, Weizhou Hou1, and Steve H. Thorne1,2
`
`Clinical
`Cancer
`Research
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Abstract
`
`Purpose: Recent data from randomized clinical trials with
`oncolytic viral therapies and with cancer immunotherapies have
`finally recapitulated the promise these platforms demonstrated in
`preclinical models. Perhaps the greatest advance with oncolytic
`virotherapy has been the appreciation of the importance of
`activation of the immune response in therapeutic activity. Mean-
`while, the understanding that blockade of immune checkpoints
`(with antibodies that block the binding of PD1 to PDL1 or CTLA4
`to B7-2) is critical for an effective antitumor immune response has
`revitalized the field of immunotherapy. The combination of
`immune activation using an oncolytic virus and blockade of
`immune checkpoints is therefore a logical next step.
`Experimental Design: Here, we explore such combinations
`and demonstrate their potential to produce enhanced responses
`in mouse tumor models. Different combinations and regimens
`were explored in immunocompetent mouse models of renal and
`
`colorectal cancer. Bioluminescence imaging and immune assays
`were used to determine the mechanisms mediating synergistic or
`antagonistic combinations.
`Results: Interaction between immune checkpoint inhibitors
`and oncolytic virotherapy was found to be complex, with correct
`selection of viral strain, antibody, and timing of the combination
`being critical for synergistic effects. Indeed, some combinations
`produced antagonistic effects and loss of therapeutic activity. A
`period of oncolytic viral replication and directed targeting of the
`immune response against the tumor were required for the most


`beneficial effects, with CD8
`and NK, but not CD4
`cells medi-
`ating the effects.
`Conclusions: These considerations will be critical in the design
`the inevitable clinical
`translation of
`these combination
`of
`approaches. Clin Cancer Res; 21(24); 5543–51. Ó2015 AACR.
`See related commentary by Slaney and Darcy, p. 5417
`
`Introduction
`The last 5 years have seen the emergence of antibody-mediated
`blockade of immune checkpoints as a key new weapon in the
`anticancer arsenal (1, 2). The anti-CTLA4 inhibitor ipilimumab
`has been approved for the treatment of melanoma (3, 4), while a
`panel of monoclonal antibodies targeting the interaction of PD1
`and PDL1 have also demonstrated promising responses in a
`succession of clinical trials (5, 6). Together, these trials have
`demonstrated the clinical need to overcome the tumor's capacity
`to shut down the T-cell response in the creation of an effective
`cancer immunotherapy.
`The field of oncolytic virotherapy has also recently demon-
`strated its potential to produce clinically effective cancer treat-
`ments, with data from several recent randomized trials resulting in
`impressive response rates (7, 8). One factor that has united the
`most successful oncolytic vectors has been the expression of an
`immune-activating transgene (GM-CSF), an indication that a key
`
`1Department of Surgery, University of Pittsburgh Cancer Institute,
`University of Pittsburgh, Pittsburgh, Pennsylvania. 2Department of
`Immunology, University of Pittsburgh Cancer Institute, University of
`Pittsburgh, Pennsylvania.
`
`Note: Supplementary data for this article are available at Clinical Cancer
`Research Online (http://clincancerres.aacrjournals.org/).
`
`Corresponding Author: Steve H. Thorne, University of Pittsburgh Cancer Institute,
`1.46e Hillman Cancer Center, 5117 Centre Avenue, Pittsburgh, PA 15213. Phone:
`412-623-4896; Fax: 412-623-2525; E-mail: ThorneSH@UPMC.edu.
`
`doi: 10.1158/1078-0432.CCR-14-2009
`Ó2015 American Association for Cancer Research.
`
`determinant of the activity of oncolytic viruses is their capacity to
`activate and target the immune response (9, 10). This has since
`been confirmed in a multitude of preclinical studies (11–13),
`such that the oncolytic virus platform might best be considered an
`immunotherapeutic.
`We have previously developed several oncolytic vectors, pri-
`marily focusing on vectors based on vaccinia virus (14–17).
`These provide several advantages as immunotherapies beyond
`their long historical use as vaccines: (i) They can induce an
`adaptive immune response raised against tumor antigens as a
`result of their selective replication within the tumor microen-
`vironment (18, 19). This in situ vaccination effect results in
`production of CTL targeting relevant tumor antigens without
`the need for any prior interrogation of the tumor. (ii) Viral
`replication within the tumor can at least transiently overcome
`localized immunosuppression, something that most traditional
`vaccine approaches fail to achieve. However, in many cases,
`once the oncolytic virus is cleared by the host immune response,
`the immunosuppressive environment is apparently restored and
`the tumor relapses. The combination of oncolytic virus and the
`blockade of
`immune checkpoint
`inhibitor therefore is an
`appealing strategy.
`Although there has been much interest in this combination,
`including the proposed clinical combinations of the oncolytic
`HSV T-Vec (Amgen) and ipilimumab (Yervoy, Bristol Myers
`Squibb) in the treatment of melanoma (Clinical Trials.gov
`NCT01740297), there have been very little supportive data
`reported to date. Here, we examine the combination of oncolytic
`vaccinia with several different immunotargeting monoclonal
`antibodies.
`
`www.aacrjournals.org
`
`5543
`
`Replimune Limited Ex. 2017 - 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/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Rojas et al.
`
`Translational Relevance
`Two of the most promising novel therapeutic platforms for
`the treatment of cancer are blockade of immune checkpoints
`and oncolytic viral therapies. Here, we look to combine these
`in preclinical mouse tumor models. The realization that inhi-
`bition of immune checkpoints is a critical need for successful
`immunotherapy and that the immune response activated by
`oncolytic viral therapies provide their most potent antitumor
`effects, means that the combination of these approaches is
`likely to result in significant clinical benefit. The enthusiasm in
`this combination is seen with the ongoing clinical combina-
`tion of the oncolytic T-Vec with ipilimumab. However, there
`has been almost no preclinical data reported to support this
`combination to date. In this article, we not only demonstrate
`that clinically relevant combinations can produce significantly
`enhanced responses in mouse tumor models, but also provide
`mechanistic insight into why some combinations are syner-
`gistic and others resulted in complete loss of therapeutic
`advantage.
`
`Materials and Methods
`Cell culture and viruses
`Renca (murine renal adenocarcinoma) cell line was obtained
`from ATCC. MC38 cell line (murine colon adenocarcinoma) was
`a kind gift from Dr. David Bartlett (University of Pittsburgh
`Cancer Institute, Pittsburrgh, PA). Cell lines were maintained in
`
`recommended culture media containing 5%–10% FBS at 37
`C,
`5% CO2. Cell lines have not been authenticated by the authors
`beyond their ability to form tumors in syngeneic mouse models.
`All recombinant vaccinia strains used in this work are derived
`from the Western Reserve (WR) strain (BEI Resources). The
`double-deleted strains vvDD and WR.B18R-.TK- (B18R- in short)
`have been described previously (15, 20). These contain deletions
`in the tk gene and in the vgf or B18R viral genes, respectively. In
`addition, both strains express the firefly luciferase gene from the
`synthetic vaccinia promoter pE/L (21), which allows monitoring
`of luciferase expression as a surrogate indicator of viral replication
`(22). Viruses were titered, manufactured, and purified as previ-
`ously described (23).
`
`Animal models
`All animal studies were approved by the University of Pitts-
`burgh Institutional Animal Care and Use Committee. C57/BL6
`and BALB/c female mice (6–8 weeks old) were purchased from
`The Jackson Laboratory. Renca or MC38 tumor cell lines were
`implanted subcutaneously at 5  105 cells per mouse into BALB/c
`or C57/BL6 mice, respectively. Oncolytic vaccinia viruses were
`injected intravenously (tail vein) at 2  108 pfu/mouse when
`tumors reached approximately 50 to 100 mm3.
`Anti-mouse CTLA4 (9D9) and anti-mouse CD25 (PC-61.5.3)
`antibodies (BioXCell) were injected intraperitoneally at 100 or
`200 mg/mouse/dose, respectively, with treatments consisting of
`3 doses each 3 days apart. Mouse IgG2b k Isotype Control
`(BioXCell) was used as a control. For depletion experiments,
`anti-mouse CD8 (2.43), anti-mouse CD4 (GK1.5), and anti-
`IFNg
`mouse
`(XMG1.2) were purchased from BioXCell,
`and anti-mouse Asialo-GM1 was purchased from Wako Pure
`
`Chemicals (Richmond, VA). Mice were injected intraperitoneally
`with 500 mg at days 1 and 2 after tumor implantation, followed by
`250 mg injection every 5 days till the end of the experiment.
`Tumor volume was monitored by caliper measurement and
`defined by V(mm3) ¼ p/6  W2  L, where W and L are the width
`and the length of the tumor, respectively. Data are expressed as
`tumor size relative to the beginning of the therapy (100%). For
`Kaplan–Meier survival curves, end point was established at 750
`mm3. Animals whose tumor size never achieved the threshold
`were included as right-censored information.
`
`Bioluminescence imaging
`Viral gene expression was determined through biolumines-
`cence imaging of luciferase expression in vivo. A dose of 4.5 mg
`of D-luciferin (GoldBio) was injected intraperitoneally per mouse
`before imaging on an IVIS2000 (PerkinElmer; 2% isoflurane).
`Images were analyzed using LivingImage software (PerkinElmer).
`
`IFNg ELISPOTs
`For ELISPOT assays, splenocytes were mixed with tumor cells or
`splenocytes from na€ve mice infected with UV-inactivated vaccinia
`virus at 5:1 ratio. Na€ve splenocytes were used as control. 96-well
`membrane filter plates (EMD Millipore) coated with 15 mg/mL of
`monoclonal anti-mouse IFNg antibody AN18 (Mabtech, Inc.)
`were used. Cells were maintained for 48 hours at 37 oC and spots
`were detected using 1 mg/mL of biotinylated anti-mouse INFg
`antibody R4-6A2-biotin (Mabtech). Plates were developed using
`an ABC kit and an AEC substrate kit for peroxidase (Vector
`Laboratories, Inc.). Specific spots were counted and analyzed using
`an ImmunoSpot Analyzer and ImmunoSpot software from CTL.
`
`Flow cytometry
`Tumors were harvested from mice and mechanically disaggre-
`gated and digested with triple enzyme mixture (Collagenase type
`IV, DNase type IV, and Hyaluronidase type V, Sigma-Aldrich)].
`Cell surface and intracellular immunostaining analyses were
`performed using a Gallios Flow Cytometer (Beckman Coulter,
`Inc.). For intracellular staining, cells were fixed and permeabilized
`using a Foxp3 Fix/Perm Buffer Set (eBioscience). Tumor-disag-
`gregated cells were stained using PE-Cy7 anti-mouse CD3 (BD
`Biosciences), eFluor450 anti-mouse NKp46, APC anti-mouse
`NKg2D, FITC anti-mouse CD4, PerCP-Cy5.5 anti-mouse CD8,
`PE anti-mouse CD25, and APC anti-mouse Foxp3 antibodies
`(eBioscience).
`
`Statistical analysis
`Standard Student t tests (two-tailed) were used throughout
`this work, except for the comparison of survival curves, where a
`log-rank test was used. In all cases, significance was achieved if
`P< 0.05.
`
`Results
`Anti-CTLA4 antibody hinders vaccinia virus replication in mice
`Mice harboring syngeneic subcutaneous mouse renal adeno-
`carcinomas (Renca cells) were injected with a single intravenous
`dose of oncolytic vaccinia virus and with three intraperitoneal
`doses of 100 mg of mouse anti-CTLA4 antibody at days 0, 3, and 6
`after virus injection. The schedule and doses of anti-CTLA4
`antibody used were determined on the basis of previously
`
`5544
`
`Clin Cancer Res; 21(24) December 15, 2015
`
`Clinical Cancer Research
`
`Replimune Limited Ex. 2017 - 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/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Oncolytic Virus Combination with Checkpoint Inhibitors
`
`published preclinical studies (24–26). Initially, we looked to
`determine the safety of the combination and the effects of injec-
`tion of anti-CTLA4 antibody on the replication of vaccinia in the
`tumors. We monitored viral luciferase transgene expression with
`bioluminescence imaging as ourselves and others have shown this
`to directly correlate with viral replication (22, 27). Anti-CTLA4
`antibody significantly reduced viral luciferase expression from
`within the tumors (at days 3 and 5 after virus injection, >5- and 40-
`fold reduction was detected; Fig. 1A). A similar depletion in viral
`replication was also observed in a second tumor model (MC38
`tumors implanted subcutaneously in C57/B6 mice, Supplemen-
`tary Fig. S1B), demonstrating that this was not a cell line- or mouse
`strain–associated effect. Reduced viral replication did, however,
`correlate with enhanced immune activation, as seen with the
`increased numbers of CTLs recognizing vaccinia epitopes detected
`in the spleens of the mice (Fig. 1B). Although increased antiviral
`CTL appeared as early as day 3 after treatment, it is likely that
`innate immune responses may also be enhanced with the com-
`bination as viral replication is reduced as early as 24 hours after
`treatment. This indicated a more robust immune response was
`raised in mice when anti-CTLA4 antibody was injected together
`with the viral therapy.
`
`Delayed administration of anti-CTLA4 antibody improves
`antitumor efficacy
`A novel schedule for oncolytic vaccinia and anti-CTLA4 anti-
`body combination was therefore designed to permit an initial
`phase of viral oncolytic activity before anti-CTLA4 antibody
`administration (Fig. 1C). Anti-CTLA4 doses were therefore
`injected at days 4, 7, and 10 after virus injection, allowing an
`initial phase of unhindered viral replication and spread within the
`tumor (Supplementary Fig. S1C). Whereas simultaneous injec-
`tion of vaccinia virus and anti-CTLA4 antibody resulted in no
`significant antitumor benefit (compared with mice treated with
`single vaccinia therapy), when we delayed administration of the
`blocking antibody until after the peak of viral replication, a greater
`than 3-fold reduction (P < 0.04) in tumor volume was observed
`(isotype control antibody had no effect on tumor growth; Fig. 1D
`and Supplementary Fig. S1A). In addition, this novel treatment
`schedule was able to significantly increase survival of mice relative
`to groups treated either with single vaccinia therapy or vaccinia
`injected concurrently with anti-CTLA4 (Fig. 1E).
`
`Combination of anti-CD25 antibody with vaccinia provided no
`therapeutic benefit
`As an alternative to immune checkpoint blockade therapy
`(anti-CTLA4), we looked to test whether other approaches that
`also target tumor immunosuppression [such as depletion of
`regulatory T cells (Treg) with anti-CD25 therapy] also synergized
`with oncolytic vaccinia therapy. We again initially monitored
`virus replication in tumors via luciferase bioluminescence imag-
`ing after anti-CD25 administration. As with anti-CTLA4 combi-
`nation, we observed a reduction in viral kinetics when antibody
`therapy began on the same day as viral treatment; however,
`differences were not significant (Fig. 2A). Furthermore, when the
`antitumor effects of vaccinia/anti-CD25 combination therapy
`were tested, neither regimen (injecting anti-CD25 antibody con-
`currently with virus or after viral replication peak) resulted in
`improved efficacy relative to single oncolytic vaccinia therapy
`(Fig. 2B and Supplementary Fig. S2B). As a further test, anti-CD25
`antibody was also added before viral therapy (Supplementary
`
`Fig. S2C), however, again no therapeutic advantage was seen (Treg
`depletion with the anti-CD25 regimen used was also confirmed;
`Supplementary Fig. S2A). Finally, a direct comparison of the
`anticancer activity of vaccinia/anti-CD25 versus vaccinia/anti-
`CTLA4 combination therapies confirmed the enhanced efficacy
`of combining oncolytic virus with blockade of CTLA4 (Fig. 2B).
`
`Immunogenicity-enhanced oncolytic vaccinia vectors improve
`synergistic effects with anti-CTLA4 antibody
`As a next step, we looked to examine the importance of the viral
`vector used in these combination approaches. Two different
`double-deleted oncolytic vaccinia viruses were compared in com-
`bination with anti-CTLA4 antibody therapy. vvDD (vgf and tk
`double-deleted vaccinia virus) has demonstrated highly tumor-
`restricted replication (28) that is equivalent in level and selectivity
`to the B18R- strain. B18R- (B18R and tk double-deleted vaccinia
`virus) also demonstrated highly tumor-restricted replication but
`this was coupled with enhanced immunogenicity relative to vvDD
`(including increased production of cytokines and chemokines
`within the tumor; ref. 29). This is due to the loss of B18R, that
`encodes a secreted type I IFN-binding protein (14). When
`both viral strains were compared for anticancer effects in combi-
`nation with anti-CTLA4 antibody (Fig. 3), B18R-/anti-CTLA4
`treatment induced a more than 3.6-fold (P < 0.009) reduction
`in tumor size at sacrifice compared with PBS treatment, while
`in this model vvDD/anti-CTLA4 combination only induced a
`1.4-fold inhibition.
`
`B18R- oncolytic vaccinia virus exhibits potent antitumor
`efficacy in optimized combination with anti-CTLA4 antibody
`therapy
`We next looked to test in more detail the most effective
`combination of viral vector (B18R-), antibody (anti-CTLA4) and
`regimen (antibody treatment beginning 4 days after viral therapy)
`determined from the previous studies.
`Mice carrying either Renca (renal adenocarcinoma) or MC38
`(colon adenocarcinoma) tumors were injected with a single
`intravenous dose of B18R- at 2  108 pfu per mouse. At days
`4, 7, and 10 after virus injection, an intraperitoneal dose of 100 mg
`of mouse anti-CTLA4 antibody was administrated. PBS or single
`therapy treatments were used as controls. At the time of sacrifice,
`combination therapy resulted in a reduction of more than 2.7-
`(P < 0.035) and 1.3-fold (P < 0.02) in Renca and MC38 tumor
`models, respectively, relative to single B18R- therapy (Fig. 4A).
`The combination induced a reduction of more than 2.8-fold
`(P < 0.04) in tumor volume compared with singe anti-CTLA4
`therapy at day 42 after treatment in Renca models. Importantly,
`B18R-/anti-CTLA4 combination therapy induced 3 of 12 com-
`plete responses in this model. For MC38 tumors, B18R-/anti-
`CTLA4 combination therapy did not produce as dramatic an
`effect, but still reduced tumor volume 1.5-fold (P < 0.045)
`compared with single anti-CTLA4 therapy, a significant improve-
`ment by day 24 after treatment.
`
`Vaccinia/anti-CTLA4 combination therapy resulted in
`enhanced systemic and tumor-specific cellular immune
`response.
`To evaluate the mechanisms driving the most effective combi-
`nation of oncolytic vaccinia and anti-CTLA4 antibody, we exam-
`ined the immune response raised against and within the tumor.
`Mice bearing Renca tumors were treated as before. Controls
`
`www.aacrjournals.org
`
`Clin Cancer Res; 21(24) December 15, 2015
`
`5545
`
`Replimune Limited Ex. 2017 - Page 3
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Rojas et al.
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Figure 1.
`Combining oncolytic vaccinia virus and anti-CTLA4 antibody therapies. A, anti-CTLA4 antibody injection reduces vaccinia virus replication in the tumor in vivo. Balb/c
`mice with subcutaneous Renca tumors (renal adenocarcinoma) were randomized and injected with a single intravenous dose of 2  108 plaque-forming units (pfu)
`per mouse of oncolytic B18R- vaccinia virus (VV). In the combination group, 100 mg of mouse anti-CTLA4 antibody was injected intraperitoneally on days
`0, 3, and 6 after virus administration. Bioluminescence imaging was used to follow viral luciferase transgene expression from within the tumor. Mean values of
`9 to 10 animals þ SD are plotted. Representative luciferase signals at day 3 after injection are also depicted (tumors are circled). B, viral/anti-CTLA4 combination
`results in increased levels of vaccinia-specific cytotoxic T cells (CTL). Mice were treated as in A, adding PBS and single therapy with anti-CTLA4 antibody as
`additional controls. At days 3 and 8 after virus injection, spleens were harvested and quantified by the IFNg ELISpot assay for vaccinia-reacting T cells. Values of
`individual mice and means  SEM of the different treatments are plotted. C, alternative schedule for vaccinia virus and anti-CTLA4 antibody combination.
`Anti-CTLA4 antibody doses were administrated at days 4, 7, and 10 after virus injection, in an approach designed to permit an initial period of viral replication.
`D, injection of anti-CTLA4 antibody after vaccinia virus replication improves therapeutic activity of combination therapy. Mice (Balb/c bearing Renca tumors)
`were treated as before or in combination with anti-CTLA4 antibody as depicted in C. Relative tumor growth and Kaplan–Meier survival curves (E) are plotted. For
`survival curves, the end point was established at a tumor volume 750 mm3. Mean values of 7 to 8 mice/group þ SE are plotted. , P < 0.05, compared
`with the VV group; f, P < 0.05, compared with the PBS group; c, P < 0.05, compared with the anti-CTLA4 group; #, P < 0.05, compared with the VVþanti-CTLA4
`day 0 group.
`
`5546
`
`Clin Cancer Res; 21(24) December 15, 2015
`
`Clinical Cancer Research
`
`Replimune Limited Ex. 2017 - Page 4
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Oncolytic Virus Combination with Checkpoint Inhibitors
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Figure 2.
`Combination of vaccinia virus with anti-CD25 antibody did not provide any therapeutic advantage. A, anti-CD25 antibody therapy effect on vaccinia virus replication.
`Balb/c mice bearing Renca tumors were injected intravenously with 2  108 pfu/mouse of oncolytic vaccinia virus (VV, strain B18R-). For the combination
`group, a dose of 200 mg of mouse anti-CD25 antibody was also injected intraperitoneally at days 0, 3, and 6 after virus administration. Viral luciferase expression
`from within the tumor was quantified at indicated time points by bioluminescence imaging. Mean values of 7 to 8 animals þ SD are plotted. Bioluminescence
`signals from one representative animal of each group at day 3 after administration are also shown (tumors are circled). B, mice (Balb/c with subcutaneous
`Renca tumors) were treated intravenously with 2  108 pfu of VV (n ¼ 10–12 per group). For combination groups, anti-CTLA4 or anti-CD25 antibodies were
`injected with 100 or 200 mg/mouse, respectively, at days 4, 7, and 10 after virus injection. PBS was injected intraperitoneally as a control. Tumor growth was
`followed by caliper measurements. Meansþ SE are plotted. , P < 0.05, compared with the PBS group; #, P < 0.05, compared with the VV group; f, P < 0.05, compared
`with the VVþ anti-CD25 day 4 group.
`
`included PBS, single B18R- therapy, or single anti-CTLA4 therapy
`(injected at days 0, 3, and 6). Mice were sacrificed at day 11 after
`virus administration and evaluated for specific CTLs in the spleen
`by the ELISpot assay and for immune cell populations in tumors
`by flow cytometry. Combination therapy was able to significantly
`increase the numbers of CTLs recognizing tumor cell antigens


`compared with any of the controls (Fig 4B). When CD3
`CD4
`populations in tumors were quantified, a significant percentage
`increase was observed after treatment with B18R-/anti-CTLA4
`combination therapy relative to any other treatment (Fig. 5A and


`C). An increase in the percentage of CD3
`CD8
`cells infiltrating
`the tumor was also observed, but appeared to be more closely
`associated with replication of the virus in the tumor (Fig. 5B and
`C), with both virus-treated groups displaying high levels of these


`cells. Finally, to ensure that the increased CD3
`CD4
`population
`infiltrating the tumors did not represent Tregs, additional staining
`for CD25 and FoxP3 was used (Fig. 5D). We observed that in the


`control group, about 40% of the CD3
`CD4
`cells present a Treg


`phenotype
`(CD25
`Foxp3
`). Anti-CTLA4 treatment barely
`reduced this percentage, but treatment with B18R- virus dropped
`amounts to 17%, and this improved further to only 13% when
`
`anti-CTLA4 was combined with oncolytic virus. Although very few
`NK or NK-T cells were detected in the tumor (<0.01% of cells), this
`number was also significantly increased only when the combi-
`nation of B18R- virus and anti-CTLA4 antibody was used in
`combination (Fig 5E and Supplementary Fig. S4).
`
`Vaccinia/anti-CTLA4 combination therapy synergistic effects


`require CD8
`T cells, NK cells, and IFNg, but not CD4
`T cells
`To define the host factors critical for the therapeutic advantage
`seen with the B18R-/anti-CTLA4 combination, viral replication
`and antitumor effect experiments were repeated in Renca tumor-


`bearing mice depleted for CD4
`T cells, CD8
`T cells, or NK cells

`(Fig. 6). It was seen that both CD8
`T cells and NK cells were
`required for the therapeutic advantage (while antitumor effects

`were maintained after depletion of CD4
`T cells; Fig. 6B). Deple-


`T cells but not NK cells or CD4
`T cells also
`tion of CD8
`significantly enhanced viral replication, indicating this cell lineage
`was responsible for both reduced viral replication and enhanced
`antitumor effects during B18R-/anti-CTLA4 combination (Fig.

`6A). Interestingly CD8
`T cells appeared responsible for reduced
`viral replication in the tumor, even at times as soon as 1 day after
`
`www.aacrjournals.org
`
`Clin Cancer Res; 21(24) December 15, 2015
`
`5547
`
`Replimune Limited Ex. 2017 - 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/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Rojas et al.
`
`Discussion
`It is evident that the blockade of immune checkpoint alone is
`rarely curative, but has the capacity to synergize with other
`therapies that selectively activate the immune response. As such,
`the realization that the immune response raised by oncolytic viral
`therapies is a critical mechanism mediating their therapeutic
`activity means that the combination of these two platforms would
`be logical and appealing (30). However, despite the fact that
`clinical trials have been proposed combining the oncolytic HSV T-
`Vec with ipilimumab, little preclinical data have been reported on
`such combinations (31).
`Here, we examine approaches to combine oncolytic vaccinia
`viruses with different monoclonal antibodies that target cancer-
`mediated immunosuppression. Significantly improved antitu-
`mor responses were demonstrated in several mouse tumor mod-
`els, providing strong support for the clinical translation of this
`approach. However, it was initially seen that careful consideration
`was needed to identify the correct combination of antibody, viral
`strain and especially in the timing of application of the different
`treatments. Incorrect combination resulted in a loss of benefit and
`potentially antagonistic effects.
`In initial studies combining anti-CTLA4 blocking antibody
`with vaccinia virus, no therapeutic benefit was seen (Fig. 1D). In
`these studies the antibody treatment was begun at the same time
`as viral inoculation, and imaging of viral luciferase transgene
`expression demonstrated that viral gene expression was reduced
`by more than 40-fold relative to virus used alone (Fig. 1A). This
`indicated that a robust antiviral immune response was being
`raised leading to premature clearance of the virus. Indeed, this
`
`Figure 3.
`Therapeutic activity of oncolytic vaccinia in combination with anti-CTLA4
`antibody is viral strain dependent. A total of 2  108 pfu of oncolytic vaccinia
`virus (B18R- or vvDD) were administrated intravenously to Balb/c mice
`bearing subcutaneous Renca tumors. At days 4, 7, and 10 after virus injection,
`a dose of 100 mg of anti-CTLA4 antibody was injected intraperitoneally.
`B18R- displayed greater inhibition of tumor growth relative to vvDD. Relative
`tumor volume after virus administration is plotted (n ¼ 12–15 mice/group þ
`SE). , P < 0.05, compared with the PBS group; f P < 0.05, compared with
`the vvDDþanti-CTLA4 day 4 group.
`

`T cells was supported
`viral treatment. The importance of CD8
`through depletion of IFNg, which also resulted in loss of thera-
`peutic advantage and enhanced viral tumor-specific replication
`(Supplementary Fig. S5). NK cells appear to be required for the
`antitumor effect, but do not limit viral replication.
`
`Figure 4.
`Optimized combination therapy
`results in synergistic anticancer
`activity. A, Renca (left) or MC38 (right)
`tumors were implanted into Balb/c or
`C57/Bl6 mice, respectively. Mice were
`injected with PBS or 2  108 pfu of
`B18R- oncolytic vaccinia virus (VV)
`through the tail vein. For the
`anti-CTLA4 group, 100 mg of
`anti-CTLA4 antibody was injected
`intraperitoneally at days 0, 3, and 6.
`For the combination group, anti-
`CTLA4 antibody doses were
`administrated at days 4, 7, and 10 after
`virus injection. Tumor volumes were
`measured, and relative tumor volume
`þ SE of the 12–15 mice/group is
`plotted. B, combination therapy
`increases cytotoxic T cells recognizing
`tumor antigens. Cellular immune
`responses to tumor cells was
`evaluated by the IFNg ELISpot assay.
`At day 11 after virus administration,
`spleens were harvested from Balb/c
`mice bearing Renca tumors and
`treated as in A. Splenocytes were
`evaluated for CTLs recognizing Renca
`cells. Values of individual mice and
`means  SEM are depicted. , P < 0.05,
`compared with the PBS group; f,
`P < 0.05, compared with the VV group;
`#, P < 0.05, compared with the
`anti-CTLA4 group.
`
`5548
`
`Clin Cancer Res; 21(24) December 15, 2015
`
`Clinical Cancer Research
`
`Replimune Limited Ex. 2017 - Page 6
`Transgene and Bioinvent International AB v. Replimune Limited
`PGR2022-00014 - U.S. Patent No. 10,947,513
`
`

`

`Oncolytic Virus Combination with Checkpoint Inhibitors
`
`Downloaded from http://aacrjournals.org/clincancerres/article-pdf/21/24/5543/2029348/5543.pdf by guest on 20 September 2022
`
`Figure 5.
`Altered T-cell repertoire in the tumor
`after vaccinia/anti-CTLA4 combination
`therapy. Balb/c mice with subcutaneous
`Renca tumors were treated as before
`(Fig. 4), and tumors were harvested at
`day 11 after virus injection and evaluated
`for lymphocyte populations by flow


`CD4
`(A)
`cytometry. Numbers of CD3


`and CD3
`CD8
`(B) cells per 200,000
`total cells are plotted. C, representative


`distributions of CD4
`and CD8

`populations within CD3
`population
`within the tumor. D, percentage of Tregs




`(CD25
`Foxp3
`) within the CD3
`CD4
`population of the tumor. Values for
`individual tumors and means þ SEM are
`plotted. E, numbers of NK cells


`
`(NKp46
`NKg2D
`CD3
`) and NK-T cells



`(NKp46
`NKg2D
`CD3
`) per 200,000
`events within the tumor. , P < 0.05,
`compared with the PBS group;
`#, P < 0.05, compared with the anti-
`CTLA4 group; f, P < 0.05, compared
`with the B18R- group.
`
`combination was also shown to result in a significant increase in
`the level of antiviral CTL (Fig. 1B) and viral replication was

`T cells or IFNg (Fig. 6A
`restored after depletion of either CD8
`and Supplementary Fig. S4A). This is potentially important as
`several groups are looking to express antibodies blocking immune
`checkpoints directly from oncolytic vectors (32). We have previ-
`ously used exogenous regulation of cytokine transgene function
`to down regulate cytokine function for a period of around 4 days
`after initial treatment (22, 33). This allowed an initial phase of
`viral oncolytic activity and unhindered replication within the
`tumor, prior to a secondary phase of immunotherapeutic activity
`that could be enhanced through subsequent stabilization of the
`cytokine function. Using a similar tactical approach, it was felt that
`addition of anti-CTLA4 antibody at later times after viral therapy
`could result in improved therapeutic activity.
`This was indeed confirmed (Fig. 1D), withinitiationof anti-CTLA4
`therapy 4 days after viral delivery found to result in significantly
`improved antitumor effects in mouse syngeneic tumor models.
`
`Several different monocl

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