l y m p h o i d c o m p a r t m e n t c y t o t o x i c s c o r e c y t ...€¦ · d a y s o n s...

1
PBS mONCR-177 PBS mONCR-177 0 2000 4000 6000 8000 HSV gB MFI **** **** PBS mONCR-177 PBS mONCR-177 0 1000 2000 3000 4000 ADPGK MFI * PBS mONCR-177 PBS mONCR-177 0 500 1000 1500 2000 2500 REPS MFI IFNg TNFa IL-2 mONCR-177 oncolytic virotherapy stimulates anti-tumor immune responses Melissa W. Hayes , Agnieszka Denslow, Jacqueline Gursha, Daniel Wambua, Lingxin Kong, Jacob Spinale, Peter Grzesik, Jennifer S. Lee, Terry Farkaly, Edward M. Kennedy, Lorena Lerner, Christophe Quéva, Brian B. Haines and Sonia Feau. Oncorus, Cambridge, MA, USA Oncorus is developing ONCR-177, an oncolytic herpes simplex virus engineered for enhanced oncolysis and antitumor immunity. ONCR-177 is replication competent in tumor cells, but its replication and neuropathic activities are attenuated in healthy cells using miRNA-dependent degradation of viral transcripts and by mutations in UL37 that prevent retrograde transport in neurons. Efficacy is enhanced by a gain of function in the fusogenic glycoprotein B to enhance viral entry and inactivation of US12 to restore antigen processing. ONCR-177 is armed with five immunostimulatory transgenes: IL-12, CCL4, FLT3LG, a PD-1 humanized antagonist VHH nanobody, and the CTLA4 blocking monoclonal antibody ipilimumab. Mechanistic evaluation is performed using mONCR-177, a mouse surrogate that expresses functionally equivalent immune stimulatory genes and the base vector, ONCR-159, which contains the same enhanced entry and inactivation, and attenuated replication and neuropathic activity, but does not carry the immunostimulatory payloads. Introduction Summary mONCR-177 induces potent in situ and abscopal efficacy, inhibits tumor growth and induces cures. Treatment with mONCR-177 increased the number of infiltrating T cells, as shown with immunohistochemistry and flow cytometry. Infiltrating cells had a higher activation status in both injected and non-injected tumor, while reducing the level of PD-1 expression. mONCR-177, compared to PBS control, elicited a more favorable CD8:T reg ratio. Potent viral antigen cytokine responses were observed with mONCR-177. mONCR-177 treatment resulted in a greater number of viral antigen and tumor antigen specific intratumoral T cells. The quality of the T cell response was enhanced in the MC38 models, with an increase in polyfunctional T cells expressing both IFNɤ and TNFα. The transcriptional profiles of treatment with mONCR-177 and combination treatment of mONCR-177+anti-PD1 resulted in a significantly improved abundance of T cells with cytotoxic capacity. The higher scores were observed in both injected and non-injected tumors and are indicative of a systemic anti-tumoral response. mONCR-177 and anti-PD1 combination treatment enhanced the efficacy of either treatment alone, improving the T cell and cytotoxicity scores. mONCR-177 Inhibits Tumor Growth and Induces Cures ONCR-177 is Uniquely Designed to Activate Multiple Arms of the Immune System ONCR-177 is delivered by intratumoral administration and is designed to elicit a potent antitumor immune response. As ONCR-177 replicates in cancer cells, it leads to cancer cells oncolysis, tumor antigens release and local production of 5 immune stimulatory transgenes. CCL4 and FLT3L promote dendritic cells recruitment and differentiation whereas IL-12 is a potent stimulator of NK cells, TH1 and cytolytic T cell responses. Antagonists of the 2 clinically validated immune checkpoints CTLA-4 and PD-1 have been added to ONCR-177 in order to facilitate T cell priming and release from immune suppression within the tumor microenvironment, respectively. Altogether these mechanisms converge to promote the generation of polyfunctional tumor-antigen specific T cells that can eradicate the injected tumor and patrol of the organism to an abscopal response in non-injected distant tumors. C57/Bl6 female mice were inoculated subcutaneously on the upper right flank and the upper left flank. Mice were divided into 3 cohorts of 5 mice per cohort. Treatment was initiated when the right flank tumor reached approximately 100 mm 3 . Mice received PBS or 6x10 6 PFU of mONCR-177, injected into a single flank tumor on day 9 and 12. Necropsy was performed on day 14, collecting the injected and non-injected tumors, and spleen from each mouse. Immunogenicity Study Plan: Bilateral MC38 tumor model, single flank intratumoral injection CD8 CD4 0 50 100 150 IT Cells/mg tumor *** ** CD8 CD4 0 20 40 60 80 Non-IT cells/mg tumor * Injected Tumor 0 5 10 15 20 %CD8+ SLEC of CD45+ * Injected Tumor 0 5 10 15 20 25 %CD8+ SLEC of CD45+ PBS mONCR-177 *** mONCR-177 Promotes Accumulation of SLEC CD8+ T cells in Injected and Non-Injected Tumors Injected Non-Injected Injected Non-Injected CD8 + (KLRG1+CD127-) were increased in the injected tumor in both tumor models when treated with mONCR-177 as well as CD4 + cells in the MC38 model. Treatment with mONCR-177 increased the number T cells in the non-injected tumor in the MC38 model. Infiltrating CD8 + T cells in both injected and non-injected tumors were predominantly short-lived effector cells. mONCR-177 Induces a Polyfunctional Viral and Tumor Antigen Response mONCR-177 Enhances the a Polyfunctional Viral Antigen Response over ONCR-159 Dissociated tumors were stimulated with viral antigen HSV gB, or tumor specific antigens, REPS1 or ADPGK for 5 hours in the presence of a golgi transport inhibitor. Cells were stained for surface phenotyping, fixed and permeabilized, then stained for intracellular cytokines. mONCR-177 induced a significantly higher expression of IFNγ in the injected and non-injected tumor in response to viral antigen. mONCR-177 also significantly increased the number of viral and tumor antigen specific IFNγ+CD8+ T cells. In response to viral antigen, mONCR-177 treatment increased the number of polyfunctional IFNγ+TNFα+CD8+ T cells in the injected tumor. *<0.05; ** <0.01; ***<0.001; ****<0.0001 Injected Non-Injected Injected Non-Injected Injected Non-Injected mONCR-177 Efficacy is Enhanced by Systemic PD-1 Blockade MC38 tumor model, mONCR-177: IT, 3x10 6 PFU, Q3Dx3; anti-PD-1 or isotype: IP, 200 mg, Q3Dx3. Gene expression analysis was performed with the Mouse PanCancer IO 360 Gene Expression Panel (Nanostring), using Day 6 MC38 tumors after 2 rounds of dosing. MC38 tumor model, mONCR-177: Intratumoral injection once tumors reached ~100mm 3 with the indicated PFU, Q3Dx3 in a single flank. Tumor growth was monitored for 20 days. Cell infiltration was evaluated by immunohistochemistry. Infiltrating CD3+ cells were quantified. The frequency of PD-1 + CD8 + T cells was similar in both tumor models. However, the level of PD-1 expression within the CD8+/PD-1+ population was lowest with mONCR-177, indicative of a less immunosuppressed tumor microenvironment. mONCR-177 improved the CD8:Treg ratio. An increase of DC1 in the spleen was observed with mONCR-159 treatment. mONCR-177 Increases the Frequency of PD-1+ TILS mONCR-177 Improves the CD8:Treg ratio mONCR-177 Increases the Number of DC1 in the Spleen Injected Non-Injected Injected Non-Injected PBS mONCR-177 0 50000 100000 150000 # of cells per spleen within the LIN- DC2 DC1 ✱✱✱ PBS mONCR-177 PBS mONCR-177 0 2000 4000 6000 8000 0 25 50 75 100 PD-1 MFI % PD-1+ CD8+ T cells % MFI ** *** PBS mONCR-177 PBS mONCR-177 0 200 400 600 800 CD8/Treg ratio ** * 0 4 8 12 16 20 24 19 20 21 22 23 Days on Study Body Weight (g) PBS mONCR-177, 3x10 4 PFU mONCR-177, 3x10 5 PFU mONCR-177, 3x10 6 PFU mONCR-177, 3x10 7 PFU C57/Bl6 female mice were inoculated subcutaneously on the upper right flank and the upper left flank. Mice were divided into 4 cohorts of 10 mice per cohort. Treatment was initiated when the right flank tumor reached approximately 100 mm 3 . Mice received PBS, 200ug/dose of an isotype control, 200ug/dose anti-PD1, 3x10 6 PFU of mONCR-177, or a combination of anti-PD1 and mONCR-177 on days 1, 4, and 7 of the study. mONCR-177 was delivered intratumorally into a single flank. Anti-PD1 was delivered intraperitoneally. Mice were monitored every 3-4 days. mONCR-177 PD-1 Combination Treatment Study Plan PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 0 2 4 6 8 10 T cell score Cell type scores-log2 Injected Non-Injected **** **** **** **** **** **** **** **** **** *** *** * PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 0 2 4 6 8 10 Cytotoxic score Cell type scores-log2 **** **** **** **** **** *** **** *** ** ** PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 -10 -5 0 5 10 Lymphoid compartment Signature scores **** **** **** **** **** *** **** **** **** ** ** ** PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 PBS IgG2a Isotype anti-PD-1 mONCR-171 mONCR-171 + anti-PD-1 -4 -2 0 2 4 6 Cytotoxicity Signature scores **** **** *** **** *** ** **** ** * ** * mONCR-177 and combination treatment elicited a more abundant T cell profile than controls or anti-PD1 alone in both injected and non-injected tumors. Further, infiltrating cells demonstrated a cytotoxic phenotype. mONCR-177 and combination treatment had the highest cytotoxic scores in both injected and non-injected tumors indicating a greater abundance of these cells. The lymphoid compartment signature measures the activity of lymphoid cells within the tumor. mONCR-177 and combination treatment demonstrated significantly higher lymphoid compartment score. The improved signature was observed for both the injected and non-injected tumors. The cytotoxicity signature measures the molecular activity of NK and CD8+ T cells. mONCR-177 and combination treatment demonstrated significantly higher cytotoxicity signature score. The improved signature was observed for both the injected and non-injected tumors. 0 20 40 60 80 0 25 50 75 100 Days on Study % Survival PBS Anti-mPD-1 mAb mONCR-171 + anti-mPD-1 mAb mONCR-171 mONCR-177 (3x107 PFU, Q3Dx3 Inhibits Bilateral MC38 Tumor Growth and is Curative ** *<0.05; ** <0.01; ***<0.001; ****<0.0001 *<0.05; ** <0.01; ***<0.001; ****<0.0001 *<0.05; ** <0.01; ***<0.001; ****<0.0001 *<0.05; ** <0.01; ***<0.001; ****<0.0001 *<0.05; ** <0.01; ***<0.001; ****<0.0001 *<0.05; ** <0.01; ***<0.001; ****<0.0001 PBS mONCR-177 PBS mONCR-177 0.0 0.5 1.0 1.5 2.0 5 10 15 20 HSV gB # of CD8 T cells/mg of tumor **** **** *** PBS mONCR-177 PBS mONCR-177 0.0 0.1 0.2 0.3 2 4 6 8 10 ADGPK # of CD8 T cells/mg of tumor **** PBS mONCR-177 PBS mONCR-177 0 2 4 6 REPS1 # of CD8 T cells/mg of tumor IFN+IL2+TNF+ IFN+IL2+TNF- IFN+IL2-TNF+ IFN+IL2-TNF- ****

Upload: others

Post on 19-Jul-2020

0 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: L y m p h o i d c o m p a r t m e n t C y t o t o x i c s c o r e C y t ...€¦ · D a y s o n S tu d y B o d y 7 W e i g h t (g) P B S m O NC R -177, 3x 10 4 P F U m O NC R -177,

PBS

mO

NCR-1

77PBS

mO

NCR-1

77

0

2000

4000

6000

8000

HS

V g

B M

FI ****

****

PBS

mO

NCR-1

77PBS

mO

NCR-1

77

0

1000

2000

3000

4000A

DP

GK

MF

I

*

PBS

mO

NCR-1

77PBS

mO

NCR-1

77

0

500

1000

1500

2000

2500

RE

PS

MF

I

IFNg

TNFa

IL-2

mONCR-177 oncolytic virotherapy stimulates anti-tumor immune responses

Melissa W. Hayes, Agnieszka Denslow, Jacqueline Gursha, Daniel Wambua, Lingxin Kong, Jacob Spinale, Peter Grzesik, Jennifer S. Lee, Terry Farkaly, Edward M. Kennedy, Lorena Lerner, Christophe Quéva, Brian B. Haines and Sonia Feau. Oncorus, Cambridge, MA, USA

Oncorus is developing ONCR-177, an oncolytic herpes simplex virus engineered for enhanced oncolysis and antitumor immunity.ONCR-177 is replication competent in tumor cells, but its replication and neuropathic activities are attenuated in healthy cellsusing miRNA-dependent degradation of viral transcripts and by mutations in UL37 that prevent retrograde transport in neurons.Efficacy is enhanced by a gain of function in the fusogenic glycoprotein B to enhance viral entry and inactivation of US12 to restoreantigen processing. ONCR-177 is armed with five immunostimulatory transgenes: IL-12, CCL4, FLT3LG, a PD-1 humanizedantagonist VHH nanobody, and the CTLA4 blocking monoclonal antibody ipilimumab. Mechanistic evaluation is performed usingmONCR-177, a mouse surrogate that expresses functionally equivalent immune stimulatory genes and the base vector, ONCR-159,which contains the same enhanced entry and inactivation, and attenuated replication and neuropathic activity, but does not carrythe immunostimulatory payloads.

Introduction

Summary

• mONCR-177 induces potent in situ and abscopalefficacy, inhibits tumor growth and inducescures.

• Treatment with mONCR-177 increased thenumber of infiltrating T cells, as shown withimmunohistochemistry and flow cytometry.Infiltrating cells had a higher activation status inboth injected and non-injected tumor, whilereducing the level of PD-1 expression.

• mONCR-177, compared to PBS control, eliciteda more favorable CD8:Treg ratio.

• Potent viral antigen cytokine responses wereobserved with mONCR-177.

• mONCR-177 treatment resulted in a greaternumber of viral antigen and tumor antigenspecific intratumoral T cells.

• The quality of the T cell response was enhancedin the MC38 models, with an increase inpolyfunctional T cells expressing both IFNɤ andTNFα.

• The transcriptional profiles of treatment withmONCR-177 and combination treatment ofmONCR-177+anti-PD1 resulted in a significantlyimproved abundance of T cells with cytotoxiccapacity. The higher scores were observed inboth injected and non-injected tumors and areindicative of a systemic anti-tumoral response.

• mONCR-177 and anti-PD1 combinationtreatment enhanced the efficacy of eithertreatment alone, improving the T cell andcytotoxicity scores.

mONCR-177 Inhibits Tumor Growth and Induces Cures

ONCR-177 is Uniquely Designed to Activate Multiple Arms of the Immune System

ONCR-177 is delivered by intratumoral administration and is designed to elicit apotent antitumor immune response. As ONCR-177 replicates in cancer cells, itleads to cancer cells oncolysis, tumor antigens release and local production of 5immune stimulatory transgenes. CCL4 and FLT3L promote dendritic cellsrecruitment and differentiation whereas IL-12 is a potent stimulator of NK cells,TH1 and cytolytic T cell responses. Antagonists of the 2 clinically validatedimmune checkpoints CTLA-4 and PD-1 have been added to ONCR-177 in order tofacilitate T cell priming and release from immune suppression within the tumormicroenvironment, respectively. Altogether these mechanisms converge topromote the generation of polyfunctional tumor-antigen specific T cells that caneradicate the injected tumor and patrol of the organism to an abscopal responsein non-injected distant tumors.

C57/Bl6 female mice were inoculated subcutaneously on the upper right flank and the upper left flank. Mice were divided into 3 cohorts of 5 mice per cohort. Treatment was initiated when the right flank tumor reached approximately 100 mm3. Mice received PBS or 6x106 PFU of mONCR-177, injected into a single flank tumor on day 9 and 12. Necropsy was performed on day 14, collecting the injected and non-injected tumors, and spleen from each mouse.

Immunogenicity Study Plan: Bilateral MC38 tumor model, single flank intratumoral injection

CD8 CD4

0

50

100

150

IT C

ells/m

g t

um

or ***

**

CD8 CD4

0

20

40

60

80

No

n-I

T c

ells/m

g t

um

or

*

Injected Tumor

0

5

10

15

20

%C

D8

+ S

LE

C o

f C

D4

5+ *

Injected Tumor

0

5

10

15

20

25

%C

D8

+ S

LE

C o

f C

D4

5+ PBS

mONCR-177

***

mONCR-177 Promotes Accumulation of SLEC CD8+ T cells in Injected and Non-Injected Tumors

Injected Non-Injected Injected Non-Injected

CD8+ (KLRG1+CD127-) were increased in the injected tumor in both tumor models when treated with mONCR-177 as well as CD4+ cells in the MC38model. Treatment with mONCR-177 increased the number T cells in the non-injected tumor in the MC38 model. Infiltrating CD8+ T cells in both injectedand non-injected tumors were predominantly short-lived effector cells.

mONCR-177 Induces a Polyfunctional Viral and Tumor Antigen Response

mONCR-177 Enhances the a Polyfunctional Viral Antigen Response over ONCR-159

Dissociated tumors were stimulated with viral antigen HSV gB, or tumor specific antigens, REPS1 or ADPGK for 5 hours in the presence of a golgi transportinhibitor. Cells were stained for surface phenotyping, fixed and permeabilized, then stained for intracellular cytokines. mONCR-177 induced a significantlyhigher expression of IFNγ in the injected and non-injected tumor in response to viral antigen. mONCR-177 also significantly increased the number of viraland tumor antigen specific IFNγ+CD8+ T cells. In response to viral antigen, mONCR-177 treatment increased the number of polyfunctionalIFNγ+TNFα+CD8+ T cells in the injected tumor. *<0.05; ** <0.01; ***<0.001; ****<0.0001

Injected Non-Injected Injected Non-Injected Injected Non-Injected

mONCR-177 Efficacy is Enhanced by Systemic PD-1 Blockade

MC38 tumor model, mONCR-177: IT, 3x106 PFU, Q3Dx3; anti-PD-1 or isotype: IP, 200 mg, Q3Dx3. Gene expression analysis was performed with the Mouse PanCancer IO 360 Gene Expression Panel (Nanostring), using Day 6 MC38 tumors after 2 rounds of dosing.

MC38 tumor model, mONCR-177: Intratumoral injection once tumors reached ~100mm3 with the indicated PFU, Q3Dx3 in a single flank. Tumor growth was monitored for 20 days. Cell infiltration was evaluated by immunohistochemistry. Infiltrating CD3+ cells were quantified.

The frequency of PD-1+ CD8+ T cells was similar in both tumor models. However, the level of PD-1 expression within the CD8+/PD-1+ population waslowest with mONCR-177, indicative of a less immunosuppressed tumor microenvironment. mONCR-177 improved the CD8:Treg ratio. An increase of DC1in the spleen was observed with mONCR-159 treatment.

mONCR-177 Increases the Frequency of PD-1+ TILS

mONCR-177 Improves the CD8:Treg ratio

mONCR-177 Increases the Number of DC1 in the Spleen

Injected Non-Injected Injected Non-Injected

PBS

mO

NCR-1

77

0

50000

100000

150000

# o

f c

ell

s p

er

sp

lee

n w

ith

in t

he

LIN

-

DC2

DC1

✱✱✱

PBS

mO

NCR-1

77PBS

mO

NCR-1

77

0

2000

4000

6000

8000

0

25

50

75

100

PD

-1 M

FI

% P

D-1

+ C

D8+

T c

ells

%

MFI

*****

PBS

mO

NCR-1

77PBS

mO

NCR-1

77

0

200

400

600

800

CD

8/T

reg

rati

o

**

*

0 4 8 12 16 20 24

19

20

21

22

23

Days on Study

Bo

dy

We

igh

t (g

) PBS

mONCR-177, 3x104 PFU

mONCR-177, 3x105 PFU

mONCR-177, 3x106 PFU

mONCR-177, 3x107 PFU

0 4 8 12 16 20 24

19

20

21

22

23

Days on Study

Bo

dy

We

igh

t (g

) PBS

mONCR-177, 3x104 PFU

mONCR-177, 3x105 PFU

mONCR-177, 3x106 PFU

mONCR-177, 3x107 PFU

C57/Bl6 female mice were inoculated subcutaneously on the upper right flank and the upper left flank. Mice were divided into 4 cohorts of 10 mice per cohort. Treatment was initiated when the right flank tumor reached approximately 100 mm3. Mice received PBS, 200ug/dose of an isotype control, 200ug/dose anti-PD1, 3x106 PFU of mONCR-177, or a combination of anti-PD1 and mONCR-177 on days 1, 4, and 7 of the study. mONCR-177 was delivered intratumorally into a single flank. Anti-PD1 was delivered intraperitoneally. Mice were monitored every 3-4 days.

mONCR-177 PD-1 Combination Treatment Study Plan

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

0

2

4

6

8

10

T cell score

Ce

ll t

yp

e s

co

res-

log

2

Injected

Non-Injected

************

************

************

*******

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

0

2

4

6

8

10

Cytotoxic score

Ce

ll t

yp

e s

co

res-

log

2

************

***********

*********

**

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

-10

-5

0

5

10

Lymphoid compartment

Sig

na

ture

sco

res

************

***********

************

******

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

PBS

IgG2a

Isoty

pe

anti-P

D-1

mON

CR-171

mON

CR-171

+ a

nti-P

D-1

-4

-2

0

2

4

6

Cytotoxicity

Sig

na

ture

sco

res

***********

********* ****

***

***

mONCR-177 and combination treatment elicited a more abundant T cell profile than controls or anti-PD1 alone in both injected and non-injected tumors.

Further, infiltrating cells demonstrated a cytotoxic phenotype. mONCR-177 and combination treatment had the highest cytotoxic scores in both injected and non-injected tumors indicating a greater abundance of these cells.

The lymphoid compartment signature measures the activity of lymphoid cells within the tumor. mONCR-177 and combination treatment demonstrated significantly higher lymphoid compartment score. The improved signature was observed for both the injected and non-injected tumors.

The cytotoxicity signature measures the molecular activity of NK and CD8+ T cells. mONCR-177 and combination treatment demonstrated significantly higher cytotoxicity signature score. The improved signature was observed for both the injected and non-injected tumors.

0 20 40 60 800

25

50

75

100

Days on Study

% S

urv

iva

l

PBS

Anti-mPD-1 mAb

mONCR-171 +

anti-mPD-1 mAb

mONCR-171

mONCR-177 (3x107 PFU, Q3Dx3 Inhibits Bilateral MC38 Tumor Growth and is Curative

**

*<0.05; ** <0.01; ***<0.001; ****<0.0001

*<0.05; ** <0.01; ***<0.001; ****<0.0001

*<0.05; ** <0.01; ***<0.001; ****<0.0001

*<0.05; ** <0.01; ***<0.001; ****<0.0001

*<0.05; ** <0.01; ***<0.001; ****<0.0001 *<0.05; ** <0.01; ***<0.001; ****<0.0001

PBS

mON

CR-177 PBS

mON

CR-177

0.0

0.5

1.0

1.5

2.0

5

10

15

20

HSV gB

# o

f C

D8 T

cells/m

g o

f tu

mo

r

****

*******

PBS

mON

CR-177 PBS

mON

CR-177

0.0

0.1

0.2

0.3

2

4

6

8

10

ADGPK

# o

f C

D8 T

cells/m

g o

f tu

mo

r

****

PBS

mON

CR-177 PBS

mON

CR-177

0

2

4

6

REPS1

# o

f C

D8 T

cells/m

g o

f tu

mo

r

IFN+IL2+TNF+

IFN+IL2+TNF-

IFN+IL2-TNF+

IFN+IL2-TNF-

****