Tumor Microenvironment Remodeling by Intratumoral Oncolytic Vaccinia Virus Enhances The Ef Ficacy of Immune-Checkpoint Blockade
Tumor Microenvironment Remodeling by Intratumoral Oncolytic Vaccinia Virus Enhances The Ef Ficacy of Immune-Checkpoint Blockade
Tumor Microenvironment Remodeling by Intratumoral Oncolytic Vaccinia Virus Enhances The Ef Ficacy of Immune-Checkpoint Blockade
Cancer
Research
Tumor Microenvironment Remodeling by
Intratumoral Oncolytic Vaccinia Virus Enhances
the Efficacy of Immune-Checkpoint Blockade
Hong Jae Chon1,2,3, Won Suk Lee1,2, Hannah Yang1,2, So Jung Kong1,2, Na Keum Lee1,2,
Eun Sang Moon4, Jiwon Choi4, Eun Chun Han2, Joo Hoon Kim1,2, Joong Bae Ahn3,
Joo Hang Kim1, and Chan Kim1,2
Abstract
Purpose: Cancer immunotherapy is a potent treatment conversion of a noninflamed tumor into an inflamed tumor.
tor. All cell lines were used within 10 passages and confirmed to
Translational Relevance be Mycoplasma free using the MycoAlert Mycoplasma Detection Kit.
Cancer immunotherapies, such as immune-checkpoint
inhibitors (ICI), have demonstrated potent therapeutic effica- Generation and quantification of virus
cy. However, many cancer patients have immunosuppressive mJX-594 (JX), provided by SillaJen, Inc., is a Western Reserve
tumors, leading to resistance against immunotherapy and strain of vaccinia virus encoding murine GM-CSF in the vaccinia
consequently limited therapeutic response. JX-594 (Pexa-vec) thymidine kinase gene locus under the control of the p7.5
is one of the most promising oncolytic virus platforms in promoter (35, 36). This virus was amplified in HeLa S3 cells
clinical development as one of the few oncolytic viruses in prior to purification. In brief, HeLa S3 cells were infected and
phase III clinical trials. Here, we show that a murine version of incubated with recombinant vaccinia virus for 3 days, collected by
JX-594 (JX) remodels the tumor microenvironment by facil- centrifugation, then homogenized and centrifuged once more.
itating the accumulation of T cells. As a result, poorly immu- The virus-containing supernatant was layered onto a 36% sucrose
nogenic tumors become sensitive to ICIs, augmenting the cushion and centrifuged at 32,900 g, and the purified viral pellet
immunotherapeutic efficacy. Of note, the triple combination was resuspended in 1 mmol/L Tris, pH 9.0. To determine the viral
therapy of JX, aPD-1, and aCTLA-4 maximizes anticancer titer, serially diluted virus in serum-free DMEM was applied onto a
immunity and induces durable regression with improved monolayer of U-2 OS cells for 2 hours, and then 1.5% carboxy-
overall survival. These findings demonstrate the potential of methylcellulose in DMEM supplemented with 2% FBS was added.
were tested using unpaired Student t tests. Survival curves were T-cell infiltration in both peritumoral and intratumoral regions
generated using the Kaplan–Meier method, and statistical differ- were also dose-dependent (Supplementary Fig. S2B). Indeed,
ences between curves were analyzed using the log-rank test. The flow-cytometric subset analysis of the lymphoid cell compart-
level of statistical significance was set at P < 0.05. ment revealed that the JX-induced increase in absolute numbers
of intratumoral CD8þ and CD4þ T cells was dose-dependent
(Supplementary Fig. S2C and S2D). Although the number of
Results CD4þFoxp3þCD25þ regulatory T cells increased following the
JX converts immunosuppressive noninflamed tumors into triple administration of JX (Supplementary Fig. S2E), the ratio of
inflamed tumors CD8þ T cells to regulatory T cells was 5.3-fold higher compared
To determine the immunomodulatory potential of the onco- with that of control treatment (Supplementary Fig. S2E), implying
lytic virus JX, we extensively examined temporal changes in TME an overall increase in T-cell effector function in the TME by JX
after single intratumoral injections of JX into the Renca tumors, treatment. Additionally, the expression of ICOS and granzyme B
which are resistant to ICIs (39). The tumoral level of JX was (GzB), which are costimulatory and T-cell activation markers,
already high at day 1, peaking at day 3, but was barely detectable at was increased in CD8þ T cells following JX treatment (Supple-
day 7 after the injection (Fig. 1A and B). Conversely, tumor vessel mentary Fig. S2F). To confirm the presence of tumor-specific T
density was markedly reduced between days 1 and 3 but was cells that were recruited into the tumor after JX treatment, IFNg
recovered at day 7 and thereafter after the injection (Fig. 1A and B; ELISPOT assays were performed with isolated TILs and spleno-
1614 Clin Cancer Res; 25(5) March 1, 2019 Clinical Cancer Research
Potentiation of Immunotherapy by Oncolytic Vaccinia Virus
Figure 1.
JX converts immunosuppressive noninflamed tumors into inflamed tumors. Renca tumors were implanted subcutaneously (s.c.) into BALB/c mice and treated
with a single intratumoral injection of 1 107 plaque-forming units (pfu) of mJX-594 (JX) when tumors reached >50 mm3. A, Representative images of Renca
tumors treated with JX. Tumor sections were stained for JX, CD31, CD8, CD11c, and PD-L1. B, Quantifications of the JXþ area, CD31þ blood vessels, CD8þ cytotoxic
T cells, CD11cþ dendritic cells, and PD-L1þ cells. , P < 0.05 versus day 0. C, Temporal changes in JX, CD8, and PD-L1 in the TME after JX treatment. D, Images
showing upregulated PD-L1 expression (red) in various cell types (green) within the TME after JX treatment. Note that the expression of PD-L1 was observed
mainly in Pan-CKþ tumor cells (arrowheads), and some CD11bþ myeloid cells (arrow) also occasionally expressed PD-L1, whereas CD3þ T cells did not. E,
NanoString immune-related gene-expression heat map. Red and green colors represent upregulated and downregulated genes, respectively. F, Volcano plot
showing changes in immune-related gene expressions in JX-treated tumors. Red line, P < 0.05. G, Comparisons of gene expressions related to inhibitory immune
checkpoints (IC), agonistic ICs, Th1 response, Th2 response, TME, and myeloid cell. Pooled data from two experiments with 5 animals per group. Values are mean
SEM. , P < 0.05 versus control. Two-tailed Student t test was used. Scale bars, 50 mm.
1616 Clin Cancer Res; 25(5) March 1, 2019 Clinical Cancer Research
Potentiation of Immunotherapy by Oncolytic Vaccinia Virus
Figure 3.
Combination of JX with aPD-1 elicits an enhanced anticancer effect with augmented infiltration of T lymphocytes into the tumor. Renca tumor–bearing mice
were treated with or without JX and aPD-1 on the indicated days (arrows). A, Comparisons of tumor growth. Mean and individual tumor growth curves over time.
B and C, Representative images (B) and comparisons (C) of CD8þ T cells, CD31þ blood vessels, activated caspase-3 (casp-3)þ apoptotic cells, and PD-L1þ cells in
the peritumoral and intratumoral regions. D, Diagram depicting the mechanism by which the immunosuppressive TME is overcome by a combination therapy of
JX and ICI. Pooled data from two experiments with 7 animals per group. Values are mean SEM. , P < 0.05 versus control; #, P < 0.05 versus JX; $, P < 0.05
versus aPD-1. ns, not significant. Two-tailed Student t test was used. Scale bars, 100 mm.
tumors (Fig. 3B and C; Supplementary Fig. S5B), implying that The therapeutic efficacy of concurrent combination therapy
PD-L1 involves an adaptive negative feedback mechanism that with ICIs and oncolytic viruses varies depending on the virus
dampens anticancer immunity after oncolytic virotherapy. administration route because viral clearance by adaptive immu-
Next, to determine whether combination therapy is effective nity may differ when the oncolytic virus is injected either system-
against distant untreated tumors as well as injected tumors, we ically (intravenous) or locally (intratumoral; refs. 22, 41). There-
treated mice carrying bilateral Renca tumors with JX and/or aPD-1 fore, we hypothesized that the administration route could affect
(Supplementary Fig. S5C). The combination therapy more the efficacy of concurrent combination therapy. To test this
potently suppressed the growth of distant untreated tumors hypothesis, we compared intravenous versus intratumoral injec-
compared with JX or aPD-1 monotherapy. tion of JX concurrently with aPD-1 (Supplementary Fig. S8A and
Therefore, our findings indicate that combining JX and ICI not S8B). Intriguingly, in tumors treated with intravenous JX, JX
only potentiates the systemic immunotherapeutic effect of JX tumoral levels were remarkably reduced with concurrent aPD-
virotherapy but also overcomes resistance against ICI monother- 1 treatment. In contrast, in tumors treated with intratumoral JX,
apy through enhanced anticancer immunity by increasing CD8þ concurrent aPD-1 treatment had almost no effect on tumoral
T-cell infiltration (Fig. 3D). levels of JX. Therefore, concurrent aPD-1 treatment seems less
We further validated our hypothesis by testing the efficacy of likely to affect JX if JX is administered via intratumoral injection.
combination treatment with anti–CTLA-4 antibody (aCTLA-4) Collectively, combination therapy with intratumoral JX injec-
and JX. Although tumor growth was modestly inhibited by either tion and systemic ICI led to an effective anticancer immunity
1618 Clin Cancer Res; 25(5) March 1, 2019 Clinical Cancer Research
Potentiation of Immunotherapy by Oncolytic Vaccinia Virus
Figure 4.
The efficacy of combination
immunotherapy with intratumoral
JX and systemic ICIs is not largely
affected by treatment schedule.
Mice were s.c. implanted with Renca
tumor cells and treated with JX plus
ICIs on various schedules. A,
Diagram depicting various
treatment schedules. Arrows
exhibited a significant reduction in overall tumor burden by triple combination immunotherapy with JX and ICIs can elicit a
48.1% and a delay in the development of palpable tumor nodules robust anticancer immune response even in a poorly immuno-
compared with control mice (Fig. 6A–D). Furthermore, triple genic spontaneous breast cancer model.
combination therapy led to a 48.1% reduction in average tumor
nodule size and better overall survival compared with other
treatments (Fig. 6E and F). Histologic analyses revealed less- Discussion
invasive carcinoma with well-preserved tumor margins in the Here, we demonstrate that combination therapy with JX and
triple combination group, indicating that triple combination ICIs is an effective therapeutic strategy for immune-resistant
effectively delays tumor progression and invasion (Fig. 6G). tumors. The combination therapy leads to an immunologic
Moreover, intratumoral recruitment of CD8þ T cells was further "boiling point" in which a cold, noninflamed tumor is sufficiently
increased by 2.0-fold in tumors treated with triple combination inflamed to enable the host immune system to eradicate tumor
therapy compared with those treated with JX monotherapy cells. The most profound effect was observed with triple immu-
(Fig. 6H). However, tumor vascular density was similar among notherapy with JX, aPD-1, and aCTLA4, which induced complete
the treatment groups (Fig. 6H), indicating that the vascular dis- regression in 40% of Renca tumors, one of the most resistant
rupting effect is not long-lasting after repeated JX injections. syngeneic tumors to immunotherapy. This strong efficacy can be
Finally, the number of hematogenous lung metastases was sig- explained by the mutually complementary cooperation of onco-
nificantly reduced in the triple combination group (Fig. 6I), lytic virus and ICIs.
indicating an effective antimetastatic action by the triple combi- JX-594 is an oncolytic virus in the most advanced stage of
nation therapy. Taken together, these results demonstrated that clinical trials and acts through various mechanisms (27, 32, 33).
Although it can rapidly induce direct oncolysis and vascular virus to prevent early shutdown of oncolytic virotherapy–
disruption in tumors, these effects are transient and mostly induced anticancer immunity (18).
diminish within 1 week of injection. Thereafter, CD8þ T cells Although ICI monotherapy revolutionized the treatment land-
extensively infiltrate the tumor to initiate anticancer immune scape of cancer, its dramatic therapeutic response is confined to a
responses. However, at the same time, tumors begin to evolve subset of patients (1, 43). This outcome gave rise to the concept of
to avoid immune-mediated elimination by upregulating immunologically "hot" or "cold" tumors: hot tumors respond
immune inhibitory checkpoint molecules such as PD-1, PD- well to ICIs because they are immunologically inflamed with TILs
L1, or CTLA-4 in the TME. Because the most potent and durable and show high expression of PD-L1, whereas cold tumors are
anticancer effects of an oncolytic virus are achieved when it is refractory to ICIs because of the paucity of CD8þ TILs and
coupled with successful induction and maintenance of antitu- immunosuppressive TME (9, 24). Therefore, current efforts are
mor immunity, it is reasonable to combine ICIs with oncolytic focused on overcoming resistance to ICIs by converting
1620 Clin Cancer Res; 25(5) March 1, 2019 Clinical Cancer Research
Potentiation of Immunotherapy by Oncolytic Vaccinia Virus
Figure 6.
The triple combination therapy delays tumor growth and metastasis in a spontaneous breast cancer model. Growth of spontaneous mammary tumors of MMTV-
PyMT mice was analyzed starting from 9 weeks after birth. Samples were harvested 13 weeks after birth. A, Diagram depicting the treatment schedule. Arrows
indicate treatment with or without intratumoral delivery of JX and systemic delivery of aPD-1 (P) and aCTLA-4 (C). B, Representative image showing gross
appearance of tumors. Dotted-line circles demarcate palpable mammary tumor nodules. C, Comparison of total tumor burden. Tumor burden was calculated by
summating the volume of every tumor nodule per mouse. D, Comparison of the number of palpable tumor nodules. E, Comparison of volume of each tumor
nodule. Each tumor nodule in MMTV-PyMT mice is plotted as an individual dot. F, Kaplan–Meier curves for overall survival. Log-rank test was used. G, H&E-stained
tumor sections showing intratumoral regions. Acinar structures of JX and JX þ P þ C groups are early, less-invasive lesions (Ea) showing the distinct boundary
with the surrounding mammary adipose tissue (Adi). Invasive ductal carcinoma regions (Ca) of Cont and P þ C have massively invaded into the surrounding
tissue and formed solid sheets of tumor cells with no remaining acinar structure. H, Representative images and comparisons of CD8þ T cells and CD31þ tumor
blood vessels in tumor. I, Representative lung sections stained with H&E and comparison of the number of metastatic colonies per lung section. Arrows indicated
metastatic foci. Unless otherwise denoted, n ¼ 8–9 for each group. Values are mean SD. , P < 0.05 versus control; #, P < 0.05 versus JX; $, P < 0.05 versus
aPD-1þ aCTLA-4. ns, not significant. Two-tailed Student t test was used in C–E, H, and I. Scale bars, 200 mm.
immunologically cold tumor to hot tumors. In this respect, our In this study, we were not able to exclude the possibility that the
result identifies JX as an ideal combination partner for ICIs. It can immunogenicity of mouse model was affected by a tumor
selectively replicate in tumor cells, destroy them, and release implantation–induced inflammatory reaction (50). Although we
tumor antigens to stimulate the host immune system. Moreover, performed every treatment 10 or 12 days after tumor implanta-
our study shows that JX can dramatically convert the TME from a tion to minimize inflammatory reaction, the level of the response
cold to hot state by inducing intratumoral inflammatory to treatment that we observed in this study may not fully reflect the
responses: induction of Th1 responses along with activation and immune reaction in human cancer. Therefore, the findings of this
recruitment of T cells, upregulation of PD-L1, and polarization of preclinical study should be confirmed in clinical trials.
myeloid cells toward M1. Intriguingly, the replication and spread Several clinical trials are ongoing to investigate the efficacy of
of oncolytic viruses are more active in cold tumors where there are JX-594 in combination with aPD-1, aCTLA-4, or aPD-L1 to
few immune cells to eliminate the virus, whereas hot tumors with target various solid cancers, including liver, kidney, and colon
ample resident TILs can induce premature clearance of virus and cancers (ClinicalTrials.gov: NCT03071094, NCT02977156,
attenuate its therapeutic effects (24). Therefore, together with the NCT03294083, and NCT03206073). Thus, we will be able to
results of this study, JX emerges as an optimal combination verify the findings of this study in a clinical setting in the near
partner for ICIs, especially for noninflamed cold tumors with future.
intrinsic resistance to immunotherapy. In conclusion, these results indicate that intratumoral injection
GM-CSF is the most commonly used therapeutic genetic pay- of JX induces a profound remodeling of the TME from cold to
References
1. Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a 3. Wolchok JD, Chan TA. Cancer: antitumour immunity gets a boost. Nature
common denominator approach to cancer therapy. Cancer Cell 2015;27: 2014;515:496–8.
450–61. 4. Ilett E, Kottke T, Thompson J, Rajani K, Zaidi S, Evgin L, et al. Prime-
2. Hegde PS, Karanikas V, Evers S. The where, the when, and the how of boost using separate oncolytic viruses in combination with check-
immune monitoring for cancer immunotherapies in the era of checkpoint point blockade improves anti-tumour therapy. Gene Ther 2017;24:
inhibition. Clin Cancer Res 2016;22:1865–74. 21–30.
1622 Clin Cancer Res; 25(5) March 1, 2019 Clinical Cancer Research
Potentiation of Immunotherapy by Oncolytic Vaccinia Virus
5. Rajani K, Parrish C, Kottke T, Thompson J, Zaidi S, Ilett L, et al. Combi- 30. Breitbach CJ, Burke J, Jonker D, Stephenson J, Haas AR, Chow LQ, et al.
nation therapy with reovirus and anti-PD-1 blockade controls tumor Intravenous delivery of a multi-mechanistic cancer-targeted oncolytic
growth through innate and adaptive immune responses. Mol Ther 2016; poxvirus in humans. Nature 2011;477:99–102.
24:166–74. 31. Breitbach CJ, De Silva NS, Falls TJ, Aladl U, Evgin L, Paterson J, et al.
6. Jung HI, Jeong D, Ji S, Ahn TS, Bae SH, Chin S, et al. Overexpression of PD- Targeting tumor vasculature with an oncolytic virus. Mol Ther 2011;19:
L1 and PD-L2 is associated with poor prognosis in patients with hepato- 886–94.
cellular carcinoma. Cancer Res Treat 2017;49:246–54. 32. Breitbach CJ, Parato K, Burke J, Hwang T-H, Bell JC, Kirn DH. Pexa-Vec
7. Topalian SL, Taube JM, Anders RA, Pardoll DM. Mechanism-driven bio- double agent engineered vaccinia: oncolytic and active immunotherapeu-
markers to guide immune checkpoint blockade in cancer therapy. tic. Curr Opin Virol 2015;13:49–54.
Nat Rev Cancer 2016;16:275–87. 33. Abou-Alfa GK, Galle PR, Chao Y, Brown KT, Heo J, Borad MJ, et al.
8. Gajewski TF. The next hurdle in cancer immunotherapy: overcoming the PHOCUS: A phase 3 randomized, open-label study comparing the onco-
non-T-cell-inflamed tumor microenvironment. Semin Oncol 2015;42: lytic immunotherapy Pexa-Vec followed by sorafenib (SOR) vs SOR in
663–71. patients with advanced hepatocellular carcinoma (HCC) without prior
9. Gajewski TF, Schreiber H, Fu YX. Innate and adaptive immune cells in the systemic therapy. J Clin Oncol 2016;34:(suppl; abstr TPS4146).
tumor microenvironment. Nat. Immunol 2013;14:1014–22. 34. Kim M, Nitschke M, Sennino B, Murer P, Schriver BJ, Bell A, et al.
10. Lechner MG, Karimi SS, Barry-Holson K, Angell TE, Murphy KA, Church CH, Amplification of oncolytic vaccinia virus widespread tumor cell killing
et al. Immunogenicity of murine solid tumor models as a defining feature of by sunitinib through multiple mechanisms. Cancer Res 2018;78:
in vivo behavior and response to immunotherapy. J. Immunother 2013;36: 922–37.
477–89. 35. Breitbach CJ, Arulanandam R, De Silva N, Thorne SH, Patt R, Daneshmand
11. De Palma M, Jain RK. CD4þ T cell activation and vascular normalization: M, et al. Oncolytic vaccinia virus disrupts tumor-associated vasculature in