Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2020 Mar 13;11(1):1395.
doi: 10.1038/s41467-020-15229-5.

An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor neoantigen-specific T cell responses

Affiliations

An engineered oncolytic virus expressing PD-L1 inhibitors activates tumor neoantigen-specific T cell responses

Guan Wang et al. Nat Commun. .

Abstract

Oncolytic viruses offer an in situ vaccination approach to activate tumor-specific T cell responses. However, the upregulation of PD-L1 expression on tumor cells and immune cells leads to tumor resistance to oncolytic immunotherapy. In this study, we generate an engineered oncolytic virus that coexpresses a PD-L1 inhibitor and GM-CSF. We find that the oncolytic virus is able to secrete the PD-L1 inhibitor that systemically binds and inhibits PD-L1 on tumor cells and immune cells. Importantly, the intratumoral injection with the oncolytic virus overcomes PD-L1-mediated immunosuppression during both the priming and effector phases, provokes systemic T cell responses against dominant and subdominant neoantigen epitopes derived from mutations, and leads to an effective rejection of both virus-injected and distant tumors. In summary, this engineered oncolytic virus is able to activate tumor neoantigen-specific T cell responses, providing a potent, individual tumor-specific oncolytic immunotherapy for cancer patients, especially those resistant to PD-1/PD-L1 blockade therapy.

PubMed Disclaimer

Conflict of interest statement

X.F.H. and J.C. are employees and shareholders of Pomona Biotechnology Corp. X.F.H. is also a shareholder of LifeSeq Limited Corp. The remaining authors declare no competing interests.

Figures

Fig. 1
Fig. 1. Generation and characterization of an oncolytic vaccinia virus coexpressing a mouse PD-L1 inhibitor and GM-CSF.
a A schematic diagram of recombinant vaccinia virus (VV) shuttle vectors that express GM-CSF or/and iPDL1 (soluble PD-1-Fc). vTK, VV thymidine kinase gene; R and L, right and left flank sequences; RFP, red fluorescent protein. b Expression and secretion of iPDL1 from infected MC38 tumor cells infected with the indicated VVs. Anti-IgG Fc (Licor 926-32210; upper) or anti-PD-1 (Biolegend 114101; lower) was used for western blot with reducing or non-reducing loading buffer. The experiment was repeated twice. c, d Serum iPDL1 and GM-CSF levels in different VV-treated MC38-bearing mice at 2 days post-virus injection c. Kinetics of iPDL1 levels in injected tumors or sera of the VV-iPDL1/GM-treated mice d. n = 3 independent samples. Data are presented as the means ± SD. The experiment was repeated twice. e Purified iPDL1 binds to PD-L1+ tumor cell. Upper panel: flow cytometric analysis of PD-L1 expression on shPD-L1/MC38 tumor cells that were transduced with PD-L1-shRNA and wild-type MC38 cells. Lower panel: shPD-L1/MC38 cells and wild-type MC38 cells were incubated with 50 μg/mL of purified iPDL1, an irrelevant MAGE3-IgG Fc fusion protein, or IgG control, followed by staining with an anti-IgG Fc for flow cytometry. f Inhibition of PD-1/PD-L1 binding by purified iPDL1 protein using ELISA. An anti-PD-L1 antibody was used as a positive control; n = 3 independent samples. g iPDL1-mediated ADCC. ADCC Reporter Bioassays were performed in triplicate wells, and the concentrations of iPDL1 protein and control IgG Fc used for this assay are indicated; n = 3 independent samples. Data presented as the means ± SD. The experiment was repeated twice. Significant differences are indicated as ***P < 0.001, or ****P < 0.0001 using two-tailed student’s t-test. h CD11c+ DC frequency in monocyte cultures in the presence of culture media of MC38 cells infected with VV-RFP, VV-GM, VV-iPDL1/GM, or GM-CSF as a positive control, and IL-4. i Viral replication in vitro; n = 3 independent samples. j Replication and biodistribution of VV after intratumor injections. Data presented as the means ± SD. The experiment was repeated twice.
Fig. 2
Fig. 2. PD-L1 inhibitors secreted from VV-iPDL1/GM-infected cells bind to PD-L1 on tumor cells and immune cells.
a MC38 tumor cells were infected with VV-RFP, VV-iPDL1/GM at an MOI = 0.5, or PBS for 24 h. The percentage of IgG Fc+ population representing iPDL1 (soluble PD-1-IgG Fc)-bound VV-infected (RFP+) or uninfected (RFP) PD-L1-expressing tumor cells was measured by flow cytometry. b MC38 tumor cells that were stimulated with IFN-γ (20 ng/mL) for 48 h were infected with the indicated VVs. PD-L1 expression of infected (RFP+) or uninfected (RFP) cells was determined by flow cytometry. cg MC38 cells were subcutaneously inoculated into the left (1 × 106) and right (5 × 105) flanks of C57BL/6 mice. When left flank tumor sizes reached ~100 mm3 (counted as day 0), the tumors of the left flank were intratumorally injected with 50 μL of PBS, VV-RFP, VV-GM, or VV-iPDL1/GM (5 × 107 pfu per tumor), or 200 μg of anti-PD-L1 antibody (clone 10F.9G2) intravenously on days 0 and 3. Two days post-second VV treatment, VV-treated (primary tumor) and untreated, distant tumors were collected, weighed and digested with collagenase type I and DNase. Tumor cell suspensions were blocked with anti-CD16/32 antibody and then stained with antibodies against CD45, CD3, CD8, CD4, CD11c, CD11b, Gr-1, FoxP3, PD-L1, and IgG Fc to assess PD-L1 expression or IgG Fc+ frequency on infected (RFP+) or uninfected (RFP) tumor cells from the treated primary tumors c or untreated distant tumors (CD45 cells) d, and PD-L1 expression on infiltrating immune cells from treated e or untreated distant tumors f, IgG Fc+ frequency on infiltrating immune cells g. Infiltrating immune cells include cytotoxic T cells (CD45+CD3+CD8+), DCs (CD45+CD11c+), myeloid-derived suppressor cell MDSCs (CD45+CD11cCD11b+Gr-1+), and Treg (CD45+CD3+CD4+FoxP3+); n = 5 mice. Significant differences are indicated as **P < 0.01, ***P < 0.001, or ****P < 0.0001 determined by two-tailed Student’s t-test.
Fig. 3
Fig. 3. Enhanced antitumor activities against primary tumors.
ac C57BL/6 mice were subcutaneously inoculated with 5 × 105 luciferase-expressing B16-F10 (B16-F10-Luc) cells. When tumor sizes reached ~100 mm3 (counted as day 0), the mice were intratumorally injected with 50 μL of VV-RFP, VV-GM, or VV-iPDL1/GM (5 × 107 pfu per tumor) or PBS at days 0, 3, and 7. Bioluminescence monitoring a, b and caliper measurement c of B16-F10-Luc cells were performed on the indicated days. Data are presented as the means ± SD (n = 5 mice). Significant differences are indicated as *P < 0.05 determined by two-tailed Student’s t-test. d, e Py230 d or MC38 e tumor volume was monitored by caliper measurement using the same treatment schedule as in ac. Data are presented as the means ± SD (n = 5 mice).
Fig. 4
Fig. 4. Enhanced antitumor activities against untreated, distant tumors.
ad Inhibition of rechallenged tumor growth. B16-F10 melanoma cells were implanted intradermally to the left flank of C57B/6 mice. When tumor sizes reached ~100 mm3 (counted as day 0), the mice were intratumorally injected with the indicated VVs on days 0, 3, and 7. Treated mice were s.c. rechallenged with B16-F10-Luc cells 30 days after the last VV injection (counted as day 0 for rechallenge data). Bioluminescence monitoring a, b and caliper measurement of B16-F10-Luc cells c were performed. Data are presented as means ± SD (n = 5 mice). d Survival curve of B16-F10 rechallenged mice. eh The volumes of rechallenged Py230 e or MC38 g tumors were monitored using a similar treatment schedule as in a, except that 5 × 105 of Py230 or MC38 tumor cells were rechallenged. Data are presented as means ± SD (n = 5 mice). *P < 0.05, ***P < 0.001 determined by two-way ANOVA. Survival curve of Py230 f and MC38 h rechallenge mice. *P < 0.05, *** P < 0.001 by two-tailed Log rank test. i CD8 T cell depletion. Surviving mice treated with VV-iPDL1/GM for the original left flank tumor implantation were rechallenged with 5 × 105 MC38 cells at right side without or with weekly i.v. injections of anti-CD8 antibody for two times. Data are presented as means ± SD (n = 5 mice). ****P < 0.01 by two-tailed repeated-measures two-way ANOVA. jl Inhibition of untreated, established tumor growth. B16-F10 melanoma cells were implanted to the left and right flanks of C57B/6 mice. The mice were intratumorally injected to the left flank tumors with indicated VVs without or with i.v. injections of anti-PD-L1 antibody on days 0, 3, and 7. j Individual curves are depicted for each tumor. Numbers indicate complete tumor regression out of total tumors in each group. k Distribution of tumor volumes determined on day 30 after virus injection; n = 10 mice. Bars represent mean values ± SD. *P < 0.05 by two-tailed Mann–Whitney U test. l Cumulative survival curves. Data are from two independent experiments. *P < 0.05; **P < 0.01; NS, not significant by two-tailed Log rank test.
Fig. 5
Fig. 5. Enhanced tumor infiltration and activation of immune cells.
A similar treatment schedule as in Fig. 2c was used, except that 5 days after the second VV injection, VV-treated MC38 tumors were harvested, weighed, and digested for preparation of single-cell suspensions followed by antibody staining against CD45, CD8, CD4, CD11c, CD11b, Gr-1, and FoxP3. ac Representative plots of the percentages of infiltrating CD45+ immune cells, DCs, MDSCs, CD4+ T cells, CD8+ T cells, and Tregs in treated tumors a. Absolute numbers of the above immune cells and CD8+ T cell/Treg ratio values in treated tumors b and distant, untreated tumors c. n = 5 mice. Data presented as the means ± SD. *P < 0.05, **P < 0.01 by two-tailed Student’s t-test. d, e Expression of IFN-γ, TNF-α, and CD 107a of tumor-infiltrating CD8+ T cells in response to restimulation with MC38 tumor lysate-pulsed DCs in the presence of Golgi-plug for 8 h were measured by intracellular staining d. e Quantitative presentation of d. n = 5 mice. Data presented as the means ± SD. *P < 0.05 by two-tailed Student’s t-test.
Fig. 6
Fig. 6. Enhanced T cell responses against dominant and subdominant tumor neoantigen epitopes.
a Enhanced T cell responses against a pool of neoantigen peptides. MC38 tumor-bearing mice were intratumorally injected with various VVs at days 0, 3, and 7. One group of C57BL/6 mice were i.v. injected with 200 μg of anti-PD-L1 antibody. Ten days later, splenocytes were cultured in the presence of a mixture of 11 neoepitope peptides (10 μg/mL/each). After 80 h of incubation, supernatants were collected for IFN-γ ELISA (right). [3H] thymidine incorporation was measured (left). The graph shows the results from three mice of each group. Data presented as the means ± SD. *P < 0.05 by two-tailed Student’s t-test. b Enhanced T cell responses against individual neoantigens. The splenocytes from VV-treated mice were cocultured with each of the 11 neoepitope peptides (100 μg/ml) as above described above. [3H] thymidine incorporation (left) and ELISA IFN-γ concentrations (right) are shown; n = 3 mice. One bar or one dot represents one mouse. Data presented as the means ± SD. *P < 0.05 by two-tailed Student’s t-test. c Enhanced T cell responses against the neoantigenic peptide 11. The splenocytes isolated from VV-treated mice were cocultured with various concentrations of the neoepitope peptide 11 as above described above. [3H] thymidine incorporation was used to analyze T cell proliferation; n = 3 mice. Data presented as the means ± SD. *P < 0.05, **P < 0.01, ***P < 0.001 by two-tailed Student’s t-test. d, e Enhanced tumor infiltration of neopeptide 4-specific T cells. Tumor cell suspensions from various VV-treated mice using the same treatment schedule as Fig. 5a were stained with the neopeptide 4 (Pep4, ASMTNMEL)-loaded, H-2Db-labeled pentamers, anti-CD45, and anti-CD8. Data are representative of five independent experiments. d Dot plots of flow cytometry; e quantification of peptide 4-pentamer+ CD8+ T cells. Data presented as the means ± SD. *P < 0.05 by two-tailed Student’s t-test. f Enhanced generation of neopeptide-specific memory T cells. Forty days after the virus injection, splenocytes were restimulated with neopeptide 4-loaded DCs in the presence of Golgi-plug followed by surface staining with anti-CD8 and intracellular staining with anti-107a, anti-IFN-γ, anti-IL-2, and anti-TNF-α.
Fig. 7
Fig. 7. Enhanced neoantigen presentation and cytolytic activity of neoantigen-specific CTLs.
a Enhanced stimulatory potency of tumor-infiltrating DCs. Tumor-infiltrating DCs from VV-treated mice were loaded with neopeptide 4, 9, or 11, and cocultured with the neoantigens-primed T cells from mice immunized with the 11 neopeptide mixture to assess IFN-γ production; n = 3 mice. Data presented as the means ± SD. *P < 0.05, ***P < 0.001 by two-tailed Student’s t-test. b Enhanced maturation of tumor-infiltrating DCs. Using a similar treatment schedule as described in Fig. 5a, cell suspensions prepared from VV-treated tumors were analyzed by flow cytometry. c Enhanced tumor infiltration of CD103+ DCs. Using the same treatment schedule as in Fig. 5a, tumor cell suspensions from VV-treated mice were analyzed by FACS; n = 5 mice. Data presented as the means ± SD. **P < 0.01 by two-tailed Student’s t-test. d Intracellular staining of IL-12 and CXCL9 of CD103+ DCs from VV-treated tumors. e qRT-PCR analysis of CXCL10 mRNA levels in CD103+ DCs isolated from VV-treated tumors; n = 5 mice. Data presented as the means ± SD. **P < 0.01 by two-tailed Student’s t-test. f Neoantigens-primed T cells proliferated more efficiently in VV-iPDL1/GM-treated mice. The neoantigens-primed T cells were labeled with 5 μM CFSE and i.v. injected into VV-treated mice. Three days later, T cell proliferation was assessed by FACS. g Enhanced stimulatory effect of VV-iPDL1/GM-infected tumor cells. MC38 tumor cells infected with VVs at MOI = 1 were cocultured with the neoantigens-primed T cells. IFN-γ production (left) and T cell proliferation (right) were measured. Data presented as the means ± SD. *P < 0.05 by two-tailed Student’s t-test. h Serum of VV-iPDL1/GM-treated mice enhanced the cytolytic activity of neoantigens-primed T cells. MC38-Luc cells were cocultured with the neoantigen-specific T cells in the presence of the sera from treated MC38-bearing mice. Cytolytic activity was calculated using luciferase emission value. Data are presented as means ± SD. **P < 0.01 by two-tailed Student’s t-test. i PD-1+ CD8+ T cells isolated from VV-treated MC38 tumors were cocultured with MC38 cells in the presence of purified iPDL1 or IgG. IFN-γ+ frequencies of PD-1+ T cells were shown from one of two independent experiments.

References

    1. Lennerz V, et al. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc. Natl Acad. Sci. USA. 2005;102:16013–16018. - PMC - PubMed
    1. Schumacher TN, Schreiber RD. Neoantigens in cancer immunotherapy. Science. 2015;348:69–74. doi: 10.1126/science.aaa4971. - DOI - PubMed
    1. Rizvi NA, et al. Cancer immunology. Mutational landscape determines sensitivity to PD-1 blockade in non-small cell lung cancer. Science. 2015;348:124–128. doi: 10.1126/science.aaa1348. - DOI - PMC - PubMed
    1. Schumacher TN, Scheper W, Kvistborg P. Cancer neoantigens. Annu. Rev. Immunol. 2018;37:173–200. doi: 10.1146/annurev-immunol-042617-053402. - DOI - PubMed
    1. Linnemann C, et al. High-throughput epitope discovery reveals frequent recognition of neo-antigens by CD4+ T cells in human melanoma. Nat. Med. 2015;21:81–85. doi: 10.1038/nm.3773. - DOI - PubMed

Publication types

MeSH terms