Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
STING LEVELS AS A BIOMARKER FOR CANCER IMMUNOTHERAPY
Document Type and Number:
WIPO Patent Application WO/2019/195658
Kind Code:
A1
Abstract:
Provided herein are STING and SPARCS genes as biomarkers for determining an effective therapy for treating cancer. Further provided are methods for treating cancer using said biomarkers.

Inventors:
BARBIE DAVID (US)
CAÑADAS ISRAEL (US)
KITAJIMA SHUNSUKE (US)
BARBIE THANH (US)
Application Number:
PCT/US2019/025959
Publication Date:
October 10, 2019
Filing Date:
April 05, 2019
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
DANA FARBER CANCER INST INC (US)
BRIGHAM & WOMENS HOSPITAL INC (US)
International Classes:
C07K14/47; C07K14/555; C12Q1/6886; G01N33/574
Domestic Patent References:
WO2016201450A22016-12-15
WO2017011920A12017-01-26
WO2017106656A12017-06-22
Foreign References:
US20160312294A12016-10-27
US20110119776A12011-05-19
US20110152206A12011-06-23
Other References:
KOTTAKIS ET AL.: "LKB1 loss links serine metabolism to DNA methylation and tumourigenesis", NATURE, vol. 539, no. 7629, 31 October 2016 (2016-10-31), pages 390 - 395, XP055642434
CORRALES ET AL.: "Innate immune signaling and regulation in cancer immunotherapy", CELL RESEARCH, vol. 27, no. 1, 16 December 2016 (2016-12-16), pages 96 - 108, XP055642436
Attorney, Agent or Firm:
BERKEY, Cristin et al. (US)
Download PDF:
Claims:
CLAIMS

What is claimed is:

Claims

1. A method for determining a treatment for cancer comprising:

(i) obtaining a biological sample obtained from a subject having cancer;

(ii) determining the level of Stimulator of IFN Genes (STING); and

(iii) selecting a therapy based on the level of STING.

2. A method for treating cancer comprising:

(i) obtaining a biological sample obtained from a subject having cancer;

(ii) determining the level of Stimulator of IFN Genes (STING);

(iii) selecting a therapy based on the level of STING; and

(iv) administering the therapy to the subject.

3. The method of claims 1 or 2, wherein the biological sample is a biopsy sample.

4. The method of any of claims 1-3, wherein the level of STING is the level of STING mRNA.

5. The method of any of claims 1-3, wherein the level of STING is the level of STING protein.

6. The method of any of the preceding claims, wherein the cancer is breast cancer, lung cancer, or kidney cancer.

7. The method of any of the preceding claims, wherein the therapy is selected from at least one of:

(i) a STING agonist;

(ii) an Immune Checkpoint Blockade (ICB) agent; and

(iii) an interferon gamma signaling agonist;

if the level of STING is above a reference value.

8. The method of claim 7, wherein the therapy comprises a STING agonist and an Immune Checkpoint Blockade (ICB) agent.

9. The method of claim 7, wherein the therapy comprises a STING agonist and an interferon gamma signaling agonist.

10. The method of any of claims 1-6, wherein the therapy is selected from at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor if the level of STING is below a reference value.

11. The method of claim 10, wherein the therapy further comprises at least one of:

(i) a STING agonist;

(ii) an Immune Checkpoint Blockade (ICB) agent; and

(iii) an interferon gamma signaling agonist.

12. The method of any of claims 7-11, wherein the STING agonist is selected from SB 11285, MK-1454, or ADU-S100.

13. The method of any of claims 7-12, wherein the Immune Checkpoint Blockade (ICB) agent is selected from anti-PDl, anti-PD-Ll, or anti-CTLA-4.

14. The method of any of claims 7-13, wherein the interferon gamma signaling agonist is selected from poly I:C or PTPN2 inhibitors.

15. The method of any of claims 7-14, wherein the DNA methyltransferase inhibitor or EZH2 inhibitor is GSK126, azacitidine, or decitabine.

16. The method of any of claims 7-15, wherein the reference is 50% or more above the control level.

17. The method of claim 16, wherein the control level is the level of STING in the same tissue type as the biological sample from a normal healthy subject.

18. A method of treating a subject having KRAS lung cancer comprising:

obtaining a biopsy sample from the subject;

determining if the sample has KRAS;LKBl+ or KRAS;LKBl mutant cancer cells; administering a STING agonist if the cancer cells are KRAS;LKBl+ cancer cells; and administering a STING agonist and at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor if the cancer cells are LKB 1 mutant cancer cells.

19. The method of claim 18, further comprising administering one or both of:

(i) an Immune Checkpoint Blockade (ICB) agent; and

(ii) an interferon gamma signaling agonist.

20. The method of claim 18 or 19, wherein the STING agonist is selected from SB 11285, MK- 1454, or ADU-S100.

21. The method of any of claims 18-20, wherein the Immune Checkpoint Blockade (ICB) agent is selected from anti-PDl, anti-PD-Ll, or anti-CTLA-4.

22. The method of any of claims 18-21, wherein the interferon gamma signaling agonist is selected from poly I:C or PTPN2 inhibitors.

23. The method of any of claims 18-22, wherein the DNA methyltransferase inhibitor or EZH2 inhibitor is GSK126, azacitidine, or decitabine.

24. A method of treating a subject having breast cancer comprising:

obtaining a biopsy sample from the subject comprising breast cancer cells;

determining if the breast cancer cells in the sample are triple negative, HER2+, or luminal B breast cancer cells;

administering a STING agonist if the breast cancer cells are triple negative, HER2+, or luminal B breast cancer cells; and administering a STING agonist and at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor if the breast cancer cells are not triple negative, HER2+, or luminal B breast cancer cells.

25. The method of claim 24, further comprising administering one or both of:

(i) an Immune Checkpoint Blockade (ICB) agent; and

(ii) an interferon gamma signaling agonist.

26. The method of claim 24 or 25, wherein the STING agonist is selected from SB 11285, MK- 1454, or ADU-S100.

27. The method of any of claims 24-26, wherein the Immune Checkpoint Blockade (ICB) agent is selected from anti-PDl, anti-PD-Ll, or anti-CTLA-4.

28. The method of any of claims 24-27, wherein the interferon gamma signaling agonist is selected from poly I:C or PTPN2 inhibitors.

29. The method of any of claims 24-28, wherein the DNA methyltransferase inhibitor or EZH2 inhibitor is GSK126, azacitidine, or decitabine.

30. A method for determining a treatment for cancer comprising:

(i) obtaining a biological sample obtained from a subject having cancer;

(ii) determining the level of one or more SPARCS genes; and

(iii) selecting a therapy based on the level of the one or more SPARCS genes.

31. A method for treating cancer comprising:

(i) obtaining a biological sample obtained from a subject having cancer;

(ii) determining the level of one or more SPARCS genes;

(iii) selecting a therapy based on the level of the one or more SPARCS genes; and

(iv) administering the therapy to the subject.

32. The method of claims 30 or 31, wherein the biological sample is a biopsy sample.

33. The method of any of claims 30-32 wherein the one or more SPARCS genes are selected from TRIM22, TRIM38, IL32, SPATS2L, EPHA3, HERC3, ADAM 19, SERPINB9, IFI44L,

F3, BEND6, AIG1, MSRB2, TNFRSF9, and ANTXR1.

34. The method of any of claims 30-33, wherein the level of the one or more SPARCS genes is the level of mRNA.

35. The method of any of claims 30-34, wherein the level of the one or more SPARCS genes is the level of protein.

36. The method of any of claims 30-35, wherein the cancer is breast cancer, lung cancer, or kidney cancer.

37. The method of any of claims 30-36, wherein the therapy is selected from at least one of:

(i) a STING agonist;

(ii) an Immune Checkpoint Blockade (ICB) agent; and

(iii) an interferon gamma signaling agonist;

if the level of the one or more SPARCS genes is above a reference value.

38. The method of claim 37, wherein the therapy comprises a STING agonist and an Immune Checkpoint Blockade (ICB) agent.

39. The method of claim 37, wherein the therapy comprises a STING agonist and an interferon gamma signaling agonist.

40. The method of any of claims 30-36, wherein the therapy is selected from at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor if the level of the one or more SPARCS genes is below a reference value.

41. The method of claim 40, wherein the therapy further comprises at least one of:

(i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and

(iii) an interferon gamma signaling agonist.

42. The method of any of claims 37-41, wherein the STING agonist is selected from SB 11285, MK-1454, or ADU-S100.

43. The method of any of claims 37-42, wherein the Immune Checkpoint Blockade (ICB) agent is selected from anti-PDl, anti-PD-Ll, or anti-CTLA-4.

44. The method of any of claims 37-43, wherein the interferon gamma signaling agonist is selected from poly I:C or PTPN2 inhibitors.

45. The method of any of claims 37-44, wherein the DNA methyltransferase inhibitor or EZH2 inhibitor is GSK126, azacitidine, or decitabine.

46. The method of any of claims 37-45, wherein the reference is 50% or more above the control level.

47. The method of claim 46, wherein the control level is the level of the one or more SPARCS genes in the same tissue type as the biological sample from a normal healthy subject.

Description:
STING LEVELS AS A BIOMARKER FOR CANCER IMMUNOTHERAPY

FEDERALLY SPONSORED RESEARCH

This invention was made with government support under R01 CA190394 awarded by the National Institutes of Health. The government has certain rights in the invention.

RELATED APPLICATIONS

This application claims the benefit under 35 U.S.C. § 119(e) of U.S. Provisional Patent Application No. 62/653,223, filed on April 5, 2018, entitled“STING LEVELS AS A

BIOMARKER FOR CANCER IMMUNOTHERAPY,” the entire contents of which is incorporated by reference herein.

BACKGROUND OF INVENTION

Tumor cell heterogeneity promotes cancer progression, as subclones can exhibit different growth properties, metastatic capacity, and drug sensitivity, and provide paracrine immune signals. One type of tumor cell heterogeneity accompanies the epithelial-mesenchymal transition (EMT). The EMT is a process by which epithelial cells lose their cell polarity and cell-cell adhesion, and gain migratory and invasive properties to become mesenchymal stem cells. EMT is essential for initiation of metastasis in cancer progression. Carcinoma cells in a primary tumor lose cell-cell adhesion mediated by E-cadherin repression and break through the basement membrane with increased invasive properties, and enter the bloodstream through intravasation. Later, when these circulating tumor cells (CTCs) exit the bloodstream to form micro-metastases, they undergo MET for clonal outgrowth at these metastatic sites. Thus, EMT and MET form the initiation and completion of the invasion-metastasis cascade. A

mesenchymal AXL/NF-kB high state associates with therapeutic resistance across multiple cancer types. Accordingly, new methods are needed for identifying and treating cancer cells according to cell state.

BRIEF SUMMARY OF INVENTION

The present disclosure is based on the surprising discovery that the level of STING and/or one or more SPARCS genes can be used as a biomarker to determine an effective cancer therapy. For example, certain cancer cells, e.g., certain tumor cells, possess high levels of STING and/or high levels of expression of one or more SPARCS genes, and that high levels of STING and/or high levels of expression of one or more SPARCS genes is a biomarker for susceptibility of cancer cells to treatment with one or more of (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist. Other cancer cells, e.g., certain tumor cells, possess low levels of STING and/or low levels of expression of one or more SPARCS genes, and that low levels of STING and/or low levels of expression of one or more SPARCS genes is a biomarker for susceptibility of cancer cells to treatment with at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor and, optionally, one or more of (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist.

In one aspect, provided herein is a method for determining a treatment for cancer. The method comprises: (i) obtaining a biological sample obtained from a subject having cancer; (ii) determining the level of Stimulator of IFN Genes (STING); and (iii) selecting a therapy based on the level of STING.

In some embodiments, the therapy is selected from at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist; if the level of STING is above a reference value. In some embodiments, the therapy comprises a STING agonist and an Immune Checkpoint Blockade (ICB) agent. In some embodiments, the therapy comprises a STING agonist and an interferon gamma signaling agonist.

In some embodiments, the therapy is selected from at least one of a DNA

methyltransferase inhibitor and an EZH2 inhibitor if the level of STING is below a reference value. In some embodiments, the therapy further comprises at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist.

In another aspect, provided herein is a method for treating cancer. The method comprises: (i) obtaining a biological sample obtained from a subject having cancer; (ii) determining the level of Stimulator of IFN Genes (STING); (iii) selecting a therapy based on the level of STING; and (iv) administering the therapy to the subject. In some embodiments, the therapy is selected from at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist; if the level of STING is above a reference value. In some embodiments, the therapy comprises a STING agonist and an Immune Checkpoint Blockade (ICB) agent. In some embodiments, the therapy comprises a STING agonist and an interferon gamma signaling agonist.

In some embodiments, the therapy is selected from at least one of a DNA

methyltransferase inhibitor and an EZH2 inhibitor if the level of STING is below a reference value. In some embodiments, the therapy further comprises at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist.

In yet another aspect, provided herein is a method of treating a subject having KRAS lung cancer. The method comprises: obtaining a biopsy sample from the subject; determining if the sample has KRAS;LKBl+ or KRAS;LKBl mutant cancer cells; administering a STING agonist if the cancer cells are KRAS;LKBl+ cancer cells; and administering a STING agonist and at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor if the cancer cells are LKB1 mutant cancer cells.

In some embodiments, the method further comprises administering one or both of: (i) an Immune Checkpoint Blockade (ICB) agent; and (ii) an interferon gamma signaling agonist.

In yet another aspect, provided herein is a method of treating a subject having breast cancer. The method comprises: obtaining a biopsy sample from the subject comprising breast cancer cells; determining if the breast cancer cells in the sample are triple negative, HER2+, or luminal B breast cancer cells; administering a STING agonist if the breast cancer cells are triple negative, HER2+, or luminal B breast cancer cells; and administering a STING agonist and at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor if the breast cancer cells are not triple negative, HER2+, or luminal B breast cancer cells.

In some embodiments, the method further comprises administering one or both of: (i) an Immune Checkpoint Blockade (ICB) agent; and (ii) an interferon gamma signaling agonist. In yet another aspect, provided herein is a method for determining a treatment for cancer. The method comprises: (i) obtaining a biological sample obtained from a subject having cancer; (ii) determining the level of one or more SPARCS genes; and (iii) selecting a therapy based on the level of the one or more SPARCS genes.

In some embodiments, the therapy is selected from at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist; if the level of one or more SPARCS genes is above a reference value. In some embodiments, the therapy comprises a STING agonist and an Immune Checkpoint Blockade (ICB) agent. In some embodiments, the therapy comprises a STING agonist and an interferon gamma signaling agonist.

In some embodiments, the therapy is selected from at least one of a DNA

methyltransferase inhibitor and an EZH2 inhibitor if the level of one or more SPARCS genes is below a reference value. In some embodiments, the therapy further comprises at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist.

In yet another aspect, provided herein is a method for treating cancer. The method comprises: (i) obtaining a biological sample obtained from a subject having cancer; (ii) determining the level of one or more SPARCS genes; (iii) selecting a therapy based on the level of the one or more SPARCS genes; and (iv) administering the therapy to the subject.

In some embodiments, the therapy is selected from at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist; if the level of one or more SPARCS genes is above a reference value. In some embodiments, the therapy comprises a STING agonist and an Immune Checkpoint Blockade (ICB) agent. In some embodiments, the therapy comprises a STING agonist and an interferon gamma signaling agonist.

In some embodiments, the therapy is selected from at least one of a DNA

methyltransferase inhibitor and an EZH2 inhibitor if the level of one or more SPARCS genes is below a reference value. In some embodiments, the therapy further comprises at least one of: (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist. In some embodiments, the level of STING is the level of STING mRNA. In some embodiments, the level of STING is the level of STING protein.

In some embodiments, the level of the one or more SPARCS genes is the level of mRNA. In some embodiments, the level of the one or more SPARCS genes is the level of protein.

In some embodiments, the one or more SPARCS genes are selected from TRIM22, TRIM38, IL32, SPATS2L, EPHA3, HERC3, ADAM 19, SERPINB9, IFI44L, F3, BEND6,

AIG1, MSRB2, TNFRSF9, and ANTXR1.

In some embodiments, the biological sample is a biopsy sample.

In some embodiments, the cancer is breast cancer, lung cancer, or kidney cancer.

In some embodiments, the STING agonist is selected from SB 11285, MK-1454, or ADU-S100.

In some embodiments, the Immune Checkpoint Blockade (ICB) agent is selected from anti-PDl, anti-PD-Ll, or anti-CTLA-4.

In some embodiments, the interferon gamma signaling agonist is selected from poly I:C or PTPN2 inhibitors.

In some embodiments, the DNA methyltransferase inhibitor or EZH2 inhibitor is GSK126, azacitidine, or decitabine.

BRIEF DESCRIPTION OF DRAWINGS

The accompanying drawings are not intended to be drawn to scale. In the drawings, each identical or nearly identical component that is illustrated in various figures is represented by a like numeral. For purposes of clarity, not every component may be labeled in every drawing. In the drawings:

Figures 1A-1F show discovery of an IFN-inducible subclass of ERVs. Figure 1A: Immunoblot of pTBKl, TBK1, IKKe, pIRF3, pERK, ERK, pAKT, AKT and tubulin levels in H69 and H69M cells after 24 or 72 h culture. Figure 1B: Fog-2 fold change cytokine/chemokine differences between H69M/H69 CM. Figure 1C: H&E and pTBKl IHC of a patient-derived SCFC brain metastasis. Scale bar indicates 20 pm. Figure 1D: Isotype control versus PD-F1 or CD44 surface expression on H69 and H69M cells ± 200 ng/mF 24 h IFNy stimulation (representative of n= 3 biological replicates). Figure 1E: Immunoblot of pTBKl, TBK1, pERK, ERK, pAKT, AKT and b-actin levels in H69, H69M, H69M-PD-Fl low , and H69M-PD-Fl high cells. Figure 1F: Log-2 fold change cytokine/chemokines differences between H69M-PD-Ll hlgh or H69M-PD-Ll low /H69 CM. Figure 1G: qRT-PCR of ERVs in H69M-PD-Ll high normalized to H69M-PD-Ll low cells, averaged from three independent experiments. Figure 1H: TRIM22 promoter and antisense orientation of MLT1C49 in the 3’UTR. Figure II: Overlap of 3’UTR antisense ERVs with H69M upregulated genes (log-2 fold change relative to H69 >2). Figure 1J: Immunoblot of STING, MAVS, pTBKl, TBK1, pIRF3, IRF3, E-Cadherin, Vimentin and b- actin levels in H69M cells after CRISPR mediated deletion of MAVS and/or STING. Figure 1K: Log-2 fold change cytokine/chemokine differences in CM from H69M cells after CRISPR mediated deletion of MAVS and/or STING compared to sgCTRL (Scramble). Figure 1L: CXCL10 Luminex absolute levels (pg/mL) in Scramble, STING KO, MAVS KO and double KO (dKO) H69M cells. Mean ± SD of duplicate samples shown. *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s / test).

Figures 2A-2N shows that SPARCS expression is inducible and triggers positive feedback amplification. Figure 2A: Isotype control versus PD-L1 or CD44 surface expression on H69AR cells ± 200 ng/mL 24 h IFNy stimulation (representative of n= 3 biological replicates). Figure 2B: Schematic of IFNy pulse treatment (200 ng/mL) of H69 or H69AR cells. Shown are control, IFNy 10 min, IFNy 30 min, IFNy 1 hr, IFNy 3 hr, from left to right, for each of H69 and H69AR. Figure 2C: qRT-PCR of MLT1C49 in H69 and H69AR cells ± 200 ng/mL IFNy pulse - 24 h chase. Mean ± SD of triplicate samples shown. Figure 2D: ATAC-seq insertion tracks of H69 and H69AR cells around TRIM22, TRIM38 and PD-L1. Differentially accessible regions indicated with arrows. Figure 2E: Immunoblot of EZH2 and b-actin in H69, H69M and H69AR cells. Figure 2F: Log-2 fold change cytokine/chemokine differences between EZH2i treated H69 cells after IFNy pulse, EZH2i treated cells, and IFNy pulsed H69 cells relative to untreated control cells. *same as H69M-PD-Ll hlgh cytokine profile in Fig. lf. Figure 2G: Log-2 fold change comparison of IFNy induced expression of SPARCS genes in EZH2i treated H69 cells versus H69AR cells. Figure 2H: qRT-PCR of 36B4 control, MLT1C49, MLT1J and MLT1A in H69AR cells + 10 min IFNy pulse - 24 h chase. RNA was treated with RNase A and

immunoprecipitated with anti-dsRNA J2 antibody, values normalized against beta-actin. Mean ± SD of triplicate samples shown. Figure 21: Immunoblot of pTBKl, TBK1, pIRF3, IRF3, pSTATl, STAT1 and b-actin levels in H69 and H69AR cells ± 200 ng/mL IFNy 10 min pulse - 24 h chase. Figure 2J: qRT-PCR of MLT1C49, IFN-b and CXCL10 in H69AR cells ± 10 min IFN-g pulse - 24 h chase. Mean ± SD of triplicate samples shown. For each graph, from left to right are control and IFN-g pulse at each of 1, 3, and 5 days. Figure 2K: qRT-PCR and ELISA of CXCL10 in sgCTRL and sgMAVS-H69AR cells 72 h following Poly(LC) transfection. Mean ± SD of triplicate samples shown. For each graph, from left to right are control and Poly(LC) transfection in each of sgCTRL and sgMAVS-H69AR cells. Figure 2L: Log-2 fold change cytokine/chemokine differences in CM between CRISPR-H69AR cells after 10 min IFNy 10 ng/mL pulse relative to sgCTRL cells (Scramble). Figure 2M: CXCL10 ELISA in Scramble, STING KO, MAVS KO and dKO H69AR CM following 10 min IFNy 10 ng/mL pulse and chase for 3 days. Mean ± SD of triplicate samples shown. From left to right are control and IFNy 10 ng/mL pulse in each of Scramble, STING KO, MAVS KO and dKO. Figure 2N: Photograph of representative excised tumors from sgCTRL and sgMAVS H69AR cells and tumor volumes measurements after 38 days of injection. Each data point represents mean ± s.e.m. tumor volumes (n=6 in sgCTRL group (top line) and n=6 in sgMAVS group (bottom line); Two-way ANOVA). *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s / test).

Figures 3A-3D show SPARCS expression across cancers. Figure 3A: ssGSEA of SPARCS signature across TCGA (n=3602 tumors) and significantly associated gene sets grouped based on biological annotations. IC = information coefficient. FDR = false discovery rate. Figure 3B: Intersection of top 1000 genes co-regulated with SPARCS in TCGA and CCLE datasets. MHC class I pathway genes in top 40 highlighted in red, EMT related genes in blue and immune evasion markers in green. Figure 3C: Distribution of high versus low SPARCS expression by TCGA cancer histology. Figure 3D: Immunoblot of AXL, MET, Vimentin, STING, MAVS and b-actin levels in cell lines with high, intermediate, or low SPARCS signature expression after 72 h culture. Figure 3E: qRT-PCR of MLT1C49, CXCL10, PD-L1 and CD44 in SPARCS high (H196, HCC44, A498, 786-0, A704, MDA-MB-231, HCC1143, 769P, and MDA-MB-468) and SPARCS low (CAKI-2, H522, T47D, MDA-MB-453, H1436, H2081, and H69) cell lines ± IFNy 10 min pulse - 24 h chase. Mean ± SD of triplicate samples shown (Mann Whitney U test).

Figures 4A-4H show that SPARCS expression is associated with adaptive and immune suppressive signatures. Figure 4A: ssGSEA of immune signatures in SPARCS hlgh and SPARCS low primary tumors across TCGA (n=3602 tumors) and ranked based on q value significance. Figure 4B: Scatterplot representing difference in SPARCS hlgh vs SPARCS low tumors of ssGSEA of immune signatures. -logio(FDR q-value) for a Student’s t test with equal variances for enrichment of ssGSEA of immune signatures in SPARCS hlgh vs SPARCS low is shown on the y-axis. Signatures more highly represented in SPARCS hlgh are shown on the right, while those more commonly represented in SPARCS low are shown on the left. Figure 4C: q value significances of ssGSEA of immune signatures in SPARCS hlgh vs SPARCS low primary tumors across TCGA. Figure 4D: TCGA RPKM values of CXCL10 and CCL2 in primary tumors grouped in SPARCS hlgh (n=50) and SPARCS low (n=50), from left to right in each graph. Figure 4E: Multiplexed immunofluorescence staining of Cytokeratin, CD8 and CD4 in KRAS mutant NSCLC human specimens used to generate PDOTS. Genomic features are shown. Scale bar indicates 100 pm. Figure 4F: Cytokine/chemokine heatmap for NSCLC PDOTS treated with Nivolumab (100 pg/mL), IFNy (200 ng/mL), or Nivolumab + IFNy plotted as Log-2 FC relative to control. Figure 4G: CXCL10 Luminex absolute levels (pg/mL) after treatment with control, Nivolumab, IFNy, or Nivolumab+IFNy, from left to right in each graph. Mean ± SD of duplicate samples shown. Figure 4H: Phase contrast images and viability quantification analysis of NSCLC PDOTS performed on Day 6 following treatment with Nivolumab (100 pg/mL), IFNy (200 ng/mL) or Nivolumab + IFNy Scale bar indicates 100 pm. *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s / test).

Figures 5A-5H show activation of innate immune signaling in mesenchymal SCLC subclones. Figure 5 A: Schematic of H69M and H69AR subclone generation. Figure 5B: Heatmap showing top signatures differentially expressed in H69M versus H69 cells. Figure 5C: Cytokine antibody arrays of 72 h CM from H69 and H69M cells. Figure 5D: Cytokine antibody arrays of 72 h CM from H187, H345, H524 (neuroendocrine phenotype) and H841, SHP77 (mesenchymal phenotype). Figure 5E: Immunoblot of pTBKl, TBK1, IKKe, pIRF3, IRF3 pERK, ERK, pAKT, AKT and tubulin levels in H841 and SHP77 cells versus H187, H345 and H524 cells after 24 or 72 h in culture. Figure 5F: Representative IHC image of weak and moderate S 172 pTBKl expression of hepatic tumor nodules obtained from a GEM model of SCLC. Figure 5G: Representative low- and high- magnification images of S 172 pTBKl heterogenous staining in metastatic lung nodules from SCLC GEMM. Figure 5H: H&E and S 172 pTBKl IHC of representative sections from lung and liver tumor nodules from another SCLC GEMM. Scale bars indicates 100 pm or 20 pm.

Figures 6A-6E show that mesenchymal subclones recruit Jurkat T cells and THP1 monocytes. Figure 6A: Schematics of co-culture of H69, H69M and H69AR spheroids with CFSE labeled Jurkat T cells or THP1 monocytes. Figures 6B and 6C: 4x and 10X phase-contrast and fluorescent imaging of H69 and H69AR in 3D microfluidic device co-cultured with CFSE labeled Jurkat T cells and THP1 monocytes. Fluorescence staining shows CFSE labeled Jurkat and THP1 cell migration into the collagen. Scale bars indicates 500 mhi (4x images) or 100 mhi (lOx images). Figure 6D: 10X phase-contrast and fluorescent imaging of H69M in 3D microfluidic device co-cultured with CFSE labeled THP1 monocytes or Jurkat T cells. Fluorescence staining shows CFSE THP1 and Jurkat cell migration into the collagen. Scale bar indicates 100 mhi. Figure 6E: Quantification of fluorescence intensity of migrated immune cells in collagen with H69, H69M and H69AR. Mean ± SD of triplicate samples shown. *p<0.05; **p<0.005; ***p<0.00l (Student’s / test).

Figures 7A-7I show the causal role of ERV sensing in the innate immune phenotype of H69M cells. Figure 7A: Schematics of PD-L1 cell sorting in H69M cells. Figure 7B: Sorting of H69M-PD-Ll hlgh and PD-Ll low subclones (phase contrast images lOx). Scale bar indicates 100 pm. Figure 7C: Phase contrast images of H69M-PD-Ll hlgh cells at different passages. Scale bar indicates 100 pm. Figure 7D: OncoPanel Assay of 300 cancer genes and 113 introns across 35 genes for mutation and copy number detection. Mutation and copy number profiles showed no significant differences. Figure 7E: Immunoblot of STING, MAVS and b-actin levels in H69, H69AR and H69M subclones. Figure 7F: 10X phase-contrast and fluorescent imaging of H69M Scramble or STING/MAVS KO cells in 3D microfluidic device co-cultured with CFSE labeled THP1 monocytes or Jurkat T cells. Fluorescence staining shows CFSE THP1 and Jurkat cell migration into the collagen. Scale bar indicates 100 pm. Figure 7G: Quantification of fluorescence intensity of migrated immune cells in collagen with H69M Scramble or STING/MAVS KO cells. Mean ± SD of triplicate samples shown. Figure 7H: Phase contrast images of H69M cells after CRISPR mediated deletion of MAVS and/or STING. Scale bar indicates 100 pm. Figure 71: qRT-PCR of E-Cadherin and Vimentin in H69M cells after CRISPR mediated deletion of MAVS and/or STING. Mean ± SD of duplicate samples shown. Scramble, STING KO, MAVS KO, and dKO are shown from left to right for each of E- Cadherin and Vimentin. *p<0.05; **p<0.005; ***p<0.00l (Student’s / test).

Figures 8A-8D show that H69AR chemoresistant cells showed an increased chromatin accessibility state specifically at SPARCS genes. Figure 8A: qRT-PCR of ERV panel in H69, H69M, H69M-PD-Ll hlgh , H69M-PD-Ll low and H69AR cells, shown from left to right for each ERV, averaged from three independent experiments. Figure 8B: qRT-PCR of TRIM22 and PD- L1 H69 and H69AR cells ± 200 ng/mL IFNy pulse - 24 h chase. Control, IFNy pulse 10 min, IFNy pulse 30 min, IFNy pulse 1 hour, and IFNy pulse 3 hour are shown from left to right in each cell type in each graph. Mean ± SD of triplicate samples shown. Figure 8C: Log-2-fold change cytokine/chemokine differences between IFNy pulsed H69AR/H69 CM. Figure 8D: ATAC-seq insertion tracks of H69 and H69AR cells around SPARCS genes, CXCL10 and CCL2. Differentially accessible regions indicated with arrows. SPARCS genes highlighted in red. *p<0.05; **p<0.005; ***p<0.00l (Student’s / test).

Figures 9A-9G show that SPARCS are silenced by EZH2 and its bidirectional transcription induces positive feedback amplification of innate immune signaling. Figure 9A: qRT-PCR of 12 different SPARCS in H69 and H69AR cells (from left to right for each gene) ± 200 ng/mL IFNy pulse - 24 h chase. Mean ± SD of triplicate samples shown. Figure 9B: qRT- PCR of 12 different SPARCS in EZH2i treated H69 cells exposed to IFNy 24 hours. Mean ± SD of triplicate samples shown. For each SPARCS gene, DMSO treated cells are shown on the left and GSK126 treated cells are shown on the right. Figure 9C: Sense/antisense transcript quantification of MLT1C49, MLT1A, MLT1J and b-actin in H69AR cells + 10 min IFNy pulse - 24 h chase. Mean ± SD of triplicate samples shown. Figure 9D: Immunoblot of pTBKl, pSTATl and b-actin levels in H69AR cells ± 200 ng/mL IFNy 10 min pulse followed by 1, 2, 3, 4, 5, 6 days’ chase. Figure 9E: qRT-PCR of MLT1A and MLT1J in H69AR cells ± 10 min IFN-g pulse - 24 h chase. In each graph, for control and IFN-g pulsed cells, days 1, 3, and 5 chase are shown from left to right. Mean ± SD of triplicate samples shown. Figure 9F: qRT-PCR of MLT1C49, IFNa, IFN/i and IFNy in H69AR cells 72 h following Poly(LC) transfection. For each of control and poly (I:C) transfection, MLT1 C49, IFNa, IRNb and IFNy in H69AR cells are shown from left to right. Mean ± SD of triplicate samples shown. Figure 9G: qRT-PCR of MLT1C49 and MLT1A in H69AR cells ± 10 min IFN-a or IFN-b pulse with indicated chase. Mean ± SD of triplicate samples shown. *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s t test).

Figures 10A-10I show positive feedback amplification of innate immune signaling induced by IFNy Figure 10A: Schematic of H69AR conditioned media experiment. Figure 10B: Immunoblot of pTBKl, TBK1, pSTATl, STAT1 and b-actin levels in H69AR cells ± 10 min IFN-g pulsed CM for 48 and 72h. Figure 10C: qRT-PCR of lFN-b (left) and CXCL10 (right) from 48h CM treated cells. Mean ± SD of triplicate samples shown. Figure 10D: qRT-PCR of ERVs from same conditions (for each of control and IFN-g pulsed CM, MLT1C49, MLT1A, and MLT1J from left to right). Mean ± SD of triplicate samples shown. Figure 10E: Immunoblot of pSTATl, STAT1 and b-actin levels in H69AR cells ± 200 ng/mL IFNy 10 min pulse followed by 24 h chase in media with DMSO, Ruxolitinib (100 nM) or MRT67307 mM). Figure 10F: qRT-PCR of CXCL10, MLT1C49, MLT1A and MLT1J in H69AR cells + 10 min IFNy - 24 h chase in media with DMSO, MRT67307 (mM) or Ruxolitinib (100 nM). For each of control and IFN-g pulsed cells, DMSO, MRT67307 and Ruxolitinib are shown from left to right. Mean ± SD of triplicate samples shown. Figure 10G: CXCL10 ELISA in H69AR cells + 10 min IFNy - 24 h chase in media with DMSO, MRT67307 (mM) or Ruxolitinib (100 nM). Mean ± SD of triplicate samples shown. For each of control and IFN-g pulsed cells, DMSO, MRT67307 and Ruxolitinib are shown from left to right. Figure 10H: Immunoblot of MAVS, STING and b-actin levels in H69AR cells after CRISPR mediated deletion of MAVS and/or STING. Figure 101: qRT-PCR of IFNfl and IFNy in sgCTRL (on left in each graph) and sgMAVS-H69AR cells (on right in each graph) 72 h following Poly(LC) transfection. Mean ± SD of triplicate samples shown. *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s / test).

Figures 11A-11B show that SPARCS positively correlates with epigenetic de-repression, TNF/NF-kB, inflammation/innate immunity, and RTK/KRAS signaling (Figure 11A) ssGSEA of SPARCS signature across CCLE (n=585 cell lines) and significantly associated gene sets grouped based on biological annotations. IC = information coefficient. FDR = false discovery rate. Figure 11B: ssGSEA of a control 3’UTR antisense ERV gene set across TCGA (n=3602 tumors) reveals lack of correlation with top SPARCS transcriptomic features.

Figures 12A-12D show that SPARCS association with multiple markers on chromosome 3p, MHC class I induction, mesenchymal state and EZH2 and SWI/SNF loss across TCGA and CCLE. Figure 12A: Heatmap showing top 20 genomic alterations associated with SPARCS signature across TCGA data set (n=3602 tumors). Figure 12B: Heatmap showing top 20 genomic alterations associated with SPARCS signature across CCLE data set (n=585 cell lines). Figure 12C: Heatmap showing top and bottom 1000 genes with expression associated with the SPARCS signature across TCGA RNA-seq datasets (n=3602 tumors). Highlighted are the positions of specific genes positively and negatively associated with the SPARCS signature. Figure 12D: Heatmap showing top and bottom 1000 genes with expression associated with the SPARCS signature across CCLE RNA-seq datasets (n=585 cell lines). Highlighted are the positions of specific genes positively and negatively associated with the SPARCS signature.

Figures 13A-13B show expression of mesenchymal markers and loss of EZH2 and multiple SWI/SNF components in SPARCS hlgh cell lines. Figure 13A: CCLE RPKM values of mesenchymal and epigenetic associated genes and dsRNA/dsDNA sensors in cell lines grouped in SPARCS hlgh (n=50) and SPARCS low (n=50). Figure 13B: Phase contrast images of SPARCS hlgh and SPARCS low cell lines. Scale bar indicates 100 mih. *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s / test).

Figures 14A-14E show that inducible surface PD-L1 correlates with high baseline CD44 expression in SPARCS hlgh cell lines and Poly (I:C) treatment of NSCLC PDOTS with de- repressed SPARCS markedly enhances CXCL10 production and sensitizes them to PD-l blockade. Figure 14A: Isotype control versus PD-L1 or CD44 surface expression on H196 (SPARCS hlgh ) and T47D (SPARCS low ) cell lines ± 200 ng/mL 24 h IFNy stimulation. Figure 14B: Classification of TCGA primary tumors based on SPARCS signature score (ssGSEA) into SPARCS hlgh (N=2225) and SPARCS low groups (N=l377). Figure 14C: Cytokine/chemokines heatmap for human NSCLC PDOTS treated with Nivolumab (100 pg/mL), Poly (I:C), or Nivolumab + Poly (I:C) plotted as Log-2 FC relative to control. Figure 14D: CXCL10 Luminex absolute levels (pg/mL) in human NSCLC PDOTS treated with control, Nivolumab (100 pg/mL), Poly (I:C), or Nivolumab + Poly (I:C), from left to right. Mean ± SD of duplicate samples shown. Figure 14E: Phase contrast images and viability quantification analysis of NSCLC PDOTS performed on Day 6 following treatment with Nivolumab (100 pg/mL), Poly (I:C), or Nivolumab + Poly (I:C). Scale bar indicates 100 pm. *p<0.05; **p<0.005; ***p<0.00l; n.s., not significant (Student’s / test).

Figures 15A and 15B. Figure 15A: qRT-PCR of STING in KL and KP cell lines. Figure 15B: Immunoblot of the indicated proteins in KL and KP cell lines. * indicates tcell lines with p53 mutation in addition to LKB1 mutation.

Figures 16A to 16C. Figure 16A: Immunoblot of the indicated proteins in KL cell lines transduced with the indicated vector. Figure 16B: Correlation between STING and DNMT1 expression in KL cell lines. Figure 16C: Immunoblot of the indicated proteins in KL cell lines transduced with the indicated vector with or without 100 nM 5-aza-2'-deoxycytidine (DAC) treatment for 7 days.

Figures 17A and 17B. Figure 17A: Immunoblot of the indicated proteins in KL cell lines transduced with the indicated vector with or without 1 ug/ml poly(dA:dT) or poly(LC) treatment for 3h. Figure 17B: ELISA of human CXCL10 levels in conditioned medium derived from KL cell lines transduced with the indicated vector with or without 1 ug/ml poly(dA:dT) or poly(LC) treatment for 24h. Figure 18 shows increased STING expression in SCLC mesenchymal subclones. Immunoblot of STING and b-actin levels in H69 (neuroendocrine) or H69AR and H69M (mesenchymal) subclones.

Figure 19 shows that EZH2 pharmacological inhibition induces STING protein expression in H69 cell line. Immunoblot of STING, histone H3K27me3 and b-actin levels in H69 cells after GSK126 (EZH2 inhibitor) treatment at the indicated concentrations and for the indicated number of days.

Figures 20A and 20B. Increased STING expression in SPARCS hlgh cell lines. Figure 20A: CCLE RPKM values of STING gene expression in cancer cell lines grouped in SPARCS hlgh (n=50) and SPARCS low (n=50). Figure 20B: Immunoblot of STING and b-actin levels in lung, breast, and kidney cancer cell lines with high, intermediate and low SPARCS signature expression.

Figure 21 shows representative STING IHC images of primary KL and KP non-small cell lung cancer (NSCLC) samples (upper panel). Insets highlight tumor cell STING expression. STING intensity in cancer cells was scored in a blinded manner (lower panel) on a scale of 0-3. IHCO: No staining, IHC1: low staining, IHC2: moderate staining, IHC3: high staining.

Figures 22A-22D show that triple negative breast cancer (TNBC) and luminal B breast cancer cell lines have elevated STING levels and TNBC cell lines are responsive to a STING agonist. Figure 22A shows increased STING expression in TNBC and luminal B breast cancer cell lines via western blot. Figure 22B shows the genotypes of the cell lines tested in Figures 22A and 22C. Figure 22C shows increased STING expression in TNBC and luminal B breast cancer cell lines via western blot. Figure 22D shows increased CXCL10 production by TNBC cell lines treated with AduroSlOO relative to controls.

DETAILED DESCRIPTION OF INVENTION

The present disclosure is based on the surprising discovery that the level of STING and/or one or more SPARCS genes can be used as a biomarker to determine an effective cancer therapy. Certain cancer cells, e.g., certain tumor cells, possess high levels of STING and that said cancer cells are susceptible to treatment with STING agonists. STING (Stimulator of interferon genes), encoded by TMEM173, has emerged as a critical regulator of the innate immune response to cytoplasmic double stranded DNA (dsDNA), and has been recognized to play an increasingly important role in cancer pathogenesis and cancer immunotherapy. STING agonists, which, in some embodiments, are analogues of the specific cyclic dinucleotide (CDN) that directly activates STING in response to cytoplasmic dsDNA, have entered the clinic in phase 1 trials both alone and in combination with immune checkpoint blockade.

Surprisingly, it has been demonstrated herein that STING levels are tightly regulated in cancer cells, and that specific cell states are associated with STING upregulation or lack thereof. Furthermore, it has been discovered that the genomic locus encoding STING ( TMEM173 ) is epigenetically regulated, and that STING expression can be induced by DNMT and EZH2 inhibition. These findings suggest that STING agonists would be most effectively deployed in specific tumor contexts with high STING levels, and that treatment with specific epigenetic inhibitors are likely to induce STING and promote sensitivity in tumors with otherwise low STING levels. Moreover, it has been found that elevated levels of STING are associated with elevated levels of one or more SPARCS genes. Accordingly, presented herein are methods for treating cancer having elevated levels of one or more SPARCS genes with STING agonists.

Elevated STING levels and/or elevated levels of one or more SPARCS genes have surprisingly been found to be associated with elevated levels of immune checkpoint proteins, e.g., PD-L1. Accordingly, presented herein are methods for treating cancer having elevated STING levels and/or elevated levels of one or more SPARCS genes with an Immune

Checkpoint Blockade (ICB) agent.

Elevated STING levels and/or elevated levels of one or more SPARCS genes have surprisingly been found to be associated with elevated levels of interferon gamma.

Accordingly, presented herein are methods for treating cancer having elevated STING levels and/or elevated levels of one or more SPARCS genes with an interferon gamma signaling agonist.

Moreover, it has been surprisingly found that expression of STING and/or expression of one or more SPARCS genes is inhibited by DNA methyltransferase and/or PRC2, e.g., EZH2. Accordingly, presented herein are methods for treating cancer cells having low levels of STING and/or low levels of one or more SPARCS genes with at least one of a DNA methyltransferase inhibitor and an EZH2 inhibitor and, optionally, one or more of (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist.

STING Biomarker Stimulator of interferon genes (STING) is a ubiquitously produced transmembrane protein encoded by the TMEM173 gene. The longest isoform of STING has 379 amino acids. STING plays a key role as a mediator of innate immune signaling. It induces the innate immune signaling in response to the detection of bacterial and viral DNA in the cytoplasm, and promotes the production of type I interferon (IFN-alpha and IFN-beta). Multiple studies have involved STING in the development of conditions including infectious diseases and certain cancers.

The STING amino acid sequence is:

MPHS S FHPS IPCPRGHG AQKA AFVFFS ACFVTFW GFGEPPEHTFR YFVFHFAS F QLGLLLN G VCS LAEELRHIHS R YRGS YWRT VRACLGCPLRRG ALLLLS IYF YY S L PN A V GPPFT WMLALLGLS Q ALNILLGLKGL AP AEIS A V CEKGNFN V AHGLA W S Y YIG YLRLILPELQ ARIRT YN QH YNNLLRG A V S QRLYILLPLDC G VPDNLS M ADP NIRFLDKLPQQTGDHAGIKDRVYSNSIYELLENGQRAGTCVLEYATPLQTLFAMS QYSQAGFSREDRLEQAKLFCRTLEDILADAPESQNNCRLIAYQEPADDSSFSLSQ E VLRHLRQEEKEE VT V GS LKT S A VPS T S TMS QEPELLIS GMEKPLPLRTDFS (SEQ ID NO: 1)

The STING amino acid sequence has GenBank Accession Number NP_938023. l. The STING nucleotide sequence has GenBank Accession Number NM_l98282.3.

In some embodiments, STING is upregulated (or is elevated, as the terms are used interchangeably) in a biological sample relative to controls. As used herein, an upregulated or elevated level includes a level that is above a control level or reference value as defined herein. An upregulated or elevated level may be, for example, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 300%, 400%, 500% or more above a control level or reference value as defined herein.

In some embodiments, a cancer, e.g., a tumor or cancer cell, with upregulated STING levels is susceptible to (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and/or (iii) an interferon gamma signaling agonist.

In some embodiments, STING is downregulated in a biological sample (or is reduced, as the terms are used interchangeably) relative to controls. As used herein, an downregulated or reduced level includes a level that is below a control level or reference value as defined herein. An downregulated or reduced level may be, for example, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 300%, 400%, 500% or more below a control level or reference value as defined herein. In some embodiments, STING levels are unchanged relative to controls.

In some embodiments, a cancer, e.g., a tumor or cancer cell, with downregulated or unchanged STING levels is susceptible to one or more of a DNA methyltransferase inhibitor and an EZH2 inhibitor, and optionally (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and/or (iii) an interferon gamma signaling agonist.

In some embodiments, a level of mRNA is upregulated. In some embodiments, a protein level is upregulated.

SPARCS Biomarkers

As is described in Example 1, Stimulated 3 Prime Antisense Retroviral Coding

Sequences (SPARCS) genes were identified as genes upregulated in cancer cells having undergone the epithelial-mesenchymal transition and having an endogenous retrovirus in the 3’ UTR. These genes are interferon-inducible genes silenced by EZH2.

In some embodiments, one or more SPARCS genes are selected from TRIM22, TRIM38, IL32, SPATS2L, EPHA3, HERC3, ADAM 19, SERPINB9, IFI44L, F3, BEND6, AIG1,

MSRB2, TNFRSF9, and ANTXR1. The GenBank Accession Nos. for the SPARCS genes are shown in Table 1 and Table 2.

In some embodiments, one or more SPARCS genes are upregulated in a biological sample (or is elevated, as the terms are used interchangeably) relative to controls. As used herein, an upregulated or elevated level includes a level that is above a control level or reference value as defined herein. An upregulated or elevated level may be, for example, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 300%, 400%, 500% or more above a control level or reference value as defined herein.

In some embodiments, the level of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 SPARCS genes is upregulated. In some embodiments, the level of all 15 SPARCS genes is upregulated.

In some embodiments, a cancer, e.g., a tumor or cancer cell, with upregulated SPARCS gene levels is susceptible to (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and/or (iii) an interferon gamma signaling agonist.

In some embodiments, one or more SPARCS genes are downregulated in a biological sample (or is reduced, as the terms are used interchangeably) relative to controls. As used herein, an downregulated or reduced level includes a level that is below a control level or reference value as defined herein. An downregulated or reduced level may be, for example, 1%, 5%, 10%, 20%, 30%, 40%, 50%, 60%, 70%, 80%, 90%, 100%, 150%, 200%, 300%, 400%, 500% or more below a control level or reference value as defined herein.

In some embodiments, the level of 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, or 14 SPARCS genes is downregulated. In some embodiments, the level of all 15 SPARCS genes is

downregulated.

In some embodiments, one or more SPARCS genes are unchanged relative to controls.

In some embodiments, a cancer, e.g., a tumor or cancer cell, with downregulated or unchanged SPARCS gene levels is susceptible to one or more of a DNA methyltransferase inhibitor and an EZH2 inhibitor, and optionally (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and/or (iii) an interferon gamma signaling agonist.

In some embodiments, a level of mRNA is upregulated. In some embodiments, a protein level is upregulated.

Table 1. GenBank Accession Numbers for Protein Sequences

Table 2. GenBank Accession Numbers for Nucleotide Sequences

Detection Methods

The methods and devices of the invention may be protein or mRNA based. Examples of protein-based assays include immunoassays (also referred to herein as immune-based assays), immunohistochemistry, flow cytometry, mass spectrometry, Western blots, Western

immunoblotting, multiplex bead-based assays, and assays involving aptamers (such as

SOMAmer™ technology) and related affinity agents. Examples of mRNA-based assays include Northern analysis, quantitative RT-PCR, microarray hybridization, and multiplex bead-based assays. These assays generally and commonly detect and measure the level of the biomarker of interest. The level of the biomarker may then be compared to a control level. Control levels will be discussed in greater detail herein. mRNA Detection

The art is familiar with various methods for analyzing mRNA levels. An exemplary quantitative RT-PCR assay may be carried out as follows: mRNA is extracted from cells in a biological sample (e.g., tumor cells) using the RNeasy kit (Qiagen). Total mRNA is used for subsequent reverse transcription using the Superscript III First-Strand Synthesis SuperMix (Invitrogen) or the Superscript VILO cDNA synthesis kit (Invitrogen). The RT reaction is used for quantitative PCR using SYBR Green PCR Master Mix and gene-specific primers, in triplicate, using an ABI 7300 Real Time PCR System.

Expression profiles of cells in a biological sample (e.g., tumor cells) can be carried out using an oligonucleotide microarray analysis. It is to be understood that such arrays may however also comprise positive and/or negative control markers such as housekeeping genes that can be used to determine if the array has been degraded and/or if the sample has been contaminated. The art is familiar with the construction of oligonucleotide arrays. See for example GeneChip Human Genome U133 Plus 2.0 Affymetrix expression array (Affymetrix). Other mRNA detection methods include multiplex detection assays well known in the art, e.g., xMAP® bead capture and detection (Luminex Corp., Austin, TX), and various oligonucleotide array assays (Illumina).

Subject. "Subject" means a mammal, such as a human, a nonhuman primate, a dog, a cat, a sheep, a horse, a cow, a pig or a goat. In an important embodiment, the mammal is a human. The subject as used herein can be an adult subject or a pediatric subject. In some embodiments, the subject has or is suspected of having cancer, e.g., any of the cancers described herein. In some embodiments, the subject is at elevated risk of developing cancer, for example, due to the presence of carcinogenic genetic mutations or exposure to carcinogens or radiation.

Biological sample. A "biological sample" from a subject can include any cellular, tissue, bone marrow, or blood sample from the subject. Any type of biological sample appropriate for conducting assays described herein can be compatible with aspects of the invention, as would be understood by one of ordinary skill in the art. In some embodiments, the biological sample is tumor tissue or a biopsy sample. mRNA Detection Binding Partners

mRNA detection binding partners include oligonucleotide or modified oligonucleotide (e.g. locked nucleic acid) probes that hybridize to a target mRNA. Probes may be designed against one or more of STING, TRIM22, TRIM38, IL32, SPATS2L, EPHA3, HERC3,

ADAM 19, SERPINB9, IFI44L, F3, BEND6, AIG1, MSRB2, TNFRSF9, and ANTXR1 or using the GenBank Accession Nos. in Table 1. Methods for designing and producing oligonucleotide probes are well known in the art (see, e.g., US Patent No. 8036835; Rimour et al. GoArrays: highly dynamic and efficient microarray probe design. Bioinformatics (2005) 21 (7): 1094-1103; and Wemersson et al. Probe selection for DNA microarrays using OligoWiz. Nat Protoc.

2007;2(l l):2677-9l).

Protein Detection

The art is familiar with various methods for analyzing protein levels. An exemplary immunoassay may be carried out as follows: A biological sample is applied to a substrate having bound to its surface biomarker- specific binding partners (i.e., immobilized biomarker- specific binding partners). The biomarker- specific binding partner (which may be referred to as a“capture ligand” because it functions to capture and immobilize the biomarker on the substrate) may be antibodies or antigen-binding antibody fragments such as Fab, F(ab)2, Fv, single chain antibodies, Fab and sFab fragments, F(ab')2, Fd fragments, scFv, and dAb fragments, although they are not so limited. Other binding partners are described herein.

Biomarkers present in the biological sample bind to the capture ligands, and the substrate is washed to remove unbound material. The substrate is then exposed to soluble biomarker- specific binding partners (which may be identical to the binding partners used to immobilize the biomarker). The soluble biomarker- specific binding partners are allowed to bind to their respective biomarkers immobilized on the substrate, and then unbound material is washed away. The substrate is then exposed to a detectable binding partner of the soluble biomarker- specific binding partner. In one embodiment, the soluble biomarker- specific binding partner is an antibody having some or all of its Fc domain. Its detectable binding partner may be an anti-Fc domain antibody. As will be appreciated by those in the art, if more than one biomarker is being detected, the assay may be configured so that the soluble biomarker- specific binding partners are all antibodies of the same isotype. In this way, a single detectable binding partner, such as an antibody specific for the common isotype, may be used to bind to all of the soluble biomarker- specific binding partners bound to the substrate. It is to be understood that the substrate may comprise capture ligands for one or more biomarkers, including two or more, three or more, four or more, five or more, etc. of the biomarkers provided by the invention.

In some instances, it may be preferable to measure biomarkers having the lowest detectable concentration. An example would be biomarkers having protein concentrations in the pg/ml range. In some instances, it may be preferable to measure, on a single substrate, biomarkers having protein concentrations that are in the same dynamic range (i.e., they are present in the biological sample in the same concentration range). Those of ordinary skill in the art will be able to devise multiplexing assays (i.e., assays that measure two or more markers) using the guidance provided herein and the knowledge in the art.

In some embodiments, the invention contemplates a substrate having a pre-determined amount of capture ligands for each biomarker. The pre-determined amount of capture ligand is may be based in part on prior measurements of biomarker levels in subjects that are STING high and STING low. The pre-determined amount of capture ligand is may be based in part on prior measurements of biomarker levels in subjects that are high for one or more SPARCS genes and low for one or more SPARCS genes. The assays may be designed such that if the subject is STING or SPARCS-positive, then one or more detectable signals appear, optionally on a biomarker-by-biomarker basis.

Other examples of protein detection methods include multiplexed immunoassays as described, e.g., in US Patent Nos. 6939720 and 8148171, and published US Patent Application No. 2008/0255766, and protein microarrays as described, e.g. in published US Patent

Application No. 2009/0088329.

Protein Detection Binding Partners

Protein detection binding partners include biomarker- specific binding partners. In some embodiments, binding partners may be antibodies. As used herein, the term“antibody” refers to a protein that includes at least one immunoglobulin variable domain or immunoglobulin variable domain sequence. For example, an antibody can include a heavy (H) chain variable region (abbreviated herein as VH), and a light (L) chain variable region (abbreviated herein as VL). In another example, an antibody includes two heavy (H) chain variable regions and two light (L) chain variable regions. The term“antibody” encompasses antigen-binding fragments of antibodies (e.g., single chain antibodies, Fab and sFab fragments, F(ab')2, Fd fragments, Fv fragments, scFv, and dAb fragments) as well as complete antibodies. Methods for making antibodies and antigen-binding fragments are well known in the art (see, e.g. Sambrook et al, "Molecular Cloning: A Laboratory Manual" (2nd Ed.), Cold Spring Harbor Laboratory Press (1989); Lewin, "Genes IV", Oxford University Press, New York, (1990), and Roitt et al., "Immunology" (2nd Ed.), Gower Medical Publishing, London, New York (1989),

W02006/040153, WO2006/122786, and W02003/002609).

In some embodiments, the binding partner for STING is the D2P2F Rabbit antibody 13647 from Cell Signaling Technology (https://www.cellsignal.com/products/primary- antibodies/sting-d2p2f-rabbit-mab/ 13647) .

Binding partners also include proteins or peptides that bind to or interact with a target biomarker, e.g. through non-covalent bonding. For example, if the biomarker is a ligand, a binding partner may be a receptor for that ligand. In another example, if the biomarker is a receptor, a binding partner may be a ligand for that receptor. In yet another example, a binding partner may be a protein or peptide known to interact with a biomarker. Methods for producing proteins are well known in the art (see, e.g. Sambrook et al, "Molecular Cloning: A Laboratory Manual" (2nd Ed.), Cold Spring Harbor Laboratory Press (1989) and Lewin, "Genes IV",

Oxford University Press, New York, (1990)) and can be used to produce binding partners such as ligands or receptors.

Binding partners also include aptamers and other related affinity agents. Aptamers include oligonucleic acid or peptide molecules that bind to a specific target molecule. Methods for producing aptamers to a target molecule are well known in the art (see, e.g., published US Patent Application No. 2009/0075834, US Patent Nos. 7435542, 7807351, and 7239742). Other examples of affinity agents include SOMAmer™ (Slow Off-rate Modified Aptamer,

SomaLogic, Boulder, CO) modified nucleic acid-based protein binding reagents.

Binding partners also include any molecule capable of demonstrating selective binding to any one of the protein targets disclosed herein, e.g., peptoids (see, e.g., Reyna J Simon et al., “Peptoids: a modular approach to drug discovery” Proceedings of the National Academy of Sciences USA, (1992), 89(20), 9367-9371; US Patent No. 5811387; and M. Muralidhar Reddy et al., Identification of candidate IgG biomarkers for Alzheimer's disease via combinatorial library screening. Cell 144, 132-142, January 7, 2011).

Detectable Labels Detectable binding partners may be directly or indirectly detectable. A directly detectable binding partner may be labeled with a detectable label such as a fluorophore. An indirectly detectable binding partner may be labeled with a moiety that acts upon (e.g., an enzyme or a catalytic domain) or is acted upon (e.g., a substrate) by another moiety in order to generate a detectable signal. These various methods and moieties for detectable labeling are known in the art.

Controls

In some embodiments, methods provided herein involve measuring a level of a biomarker in a biological sample and comparing the biomarker level to a control level in order to characterize the level of STING or SPARCS expression in a subject. The control level is a level of the same biomarker in a control tissue, control subject, or a population of control subjects.

The“control” may be (or may be derived from) a normal subject (or normal subjects). Normal subjects, as used herein, refer to subjects that are apparently healthy and show no cancer symptoms. The control population may therefore be a population of normal subjects. In some embodiments, the control is from a normal healthy subject or subjects and is from the same tissue type as the biological sample.

In other instances, the control may be (or may be derived from) an ill subject (or ill subjects) that presents with one or more cancer without elevated or reduced STING or SPARCS expression.

In other instances, the control may be (or may be derived from) an ill subject (or ill subjects) that presents with one or more cancer with elevated or reduced STING or SPARCS expression.

In some embodiments, the control may be non-cancerous tissue from the subject being tested.

In still other instances, the control may be a biomarker level from a population of subjects regardless of whether they manifest or do not manifest cancer-like symptoms (e.g., a subset of the general population).

In any of the above embodiments, the control may be of the same tissue type as the biological sample.

It is to be understood however that the methods provided herein do not require that a control level be measured every time a subject is tested. In some embodiments, a control level be measured when a subject is tested. In other embodiments, it is contemplated that control levels of biomarkers are obtained and recorded and that any test level is compared to such a pre determined level. Such pre-determined control levels may also be referred to herein as reference values which are discussed in greater detail herein.

Reference Values

A reference value describes a measurement value for a biomarker that aids in identifying a subject with a cancer susceptible to treatment with agents described herein.

The reference value may be calculated from control levels of each biomarker as measured from a control subject or population of subjects, or from non-cancerous tissue.

In some embodiments, the reference value for STING is the same as or 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 50%, 100%, or more above the control level. In some embodiments, the reference value for STING is 1%, 2%, 3%, 4%, or 5% below the control level. In some embodiments, the reference value for the one or more SPARCS genes is the same as or 1%, 2%, 3%, 4%, 5%, 10%, 15%, 20%, 50%, 100%, or more above the control level. In some

embodiments, the reference value for the one or more SPARCS genes is 1%, 2%, 3%, 4%, or 5% below the control level.

The foregoing reference values are exemplary and it is to be understood that one of ordinary skill in the art is able to determine a control level based on the teachings provided herein and thereby establish new reference values that may be used in the methods provided herein.

Treatment

In some embodiments, a subject with a STING level or a level of one or more SPARCS genes above a reference value is treated with one or more of (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist. In some embodiments, a subject with a STING level or a level of one or more SPARCS genes below a reference value is treated with one or more of a DNA methyltransferase inhibitor and an EZH2 inhibitor and optionally one or more of (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist.

In some embodiments, the subject to be treated by the methods described herein is human. In some embodiments, a human subject who needs the treatment may be a human patient having, at risk for, or suspected of having cancer. A subject having cancer can be identified by routine medical examination, e.g., laboratory tests, functional tests, biopsy, CT scans, or ultrasounds. A subject suspected of having cancer might show one or more symptoms of the disorder. A subject at risk for cancer can be a subject having one or more of the risk factors for that disorder. For example, risk factors associated with cancer include (a) hereditary cancer, (b) age, and (c) family history of cancer.

Cancers include but are not limited to: Oral: buccal cavity, lip, tongue, mouth, pharynx; Cardiac: sarcoma (angiosarcoma, fibrosarcoma, rhabdomyosarcoma, liposarcoma), myxoma, rhabdomyoma, fibroma, lipoma and teratoma; Lung: non-small cell lung cancer (NSCLC), small cell lung cancer, bronchogenic carcinoma (squamous cell or epidermoid, undifferentiated small cell, undifferentiated large cell, adenocarcinoma), alveolar (bronchiolar) carcinoma, bronchial adenoma, sarcoma, lymphoma, chondromatous hamartoma, mesothelioma; Gastrointestinal: esophagus (squamous cell carcinoma, larynx, adenocarcinoma, leiomyosarcoma, lymphoma), stomach (carcinoma, lymphoma, leiomyosarcoma), pancreas (ductal adenocarcinoma, insulinoma, glucagonoma, gastrinoma, carcinoid tumors, vipoma), small bowel or small intestines (adenocarcinoma, lymphoma, carcinoid tumors, Karposi's sarcoma, leiomyoma, hemangioma, lipoma, neurofibroma, fibroma), large bowel or large intestines (adenocarcinoma, tubular adenoma, villous adenoma, hamartoma, leiomyoma), rectal, colon, colon-rectum, colorectal; Genitourinary tract: kidney (adenocarcinoma, Wilm's tumor [nephroblastoma], lymphoma, leukemia), bladder and urethra (squamous cell carcinoma, transitional cell carcinoma, adenocarcinoma), prostate (adenocarcinoma, sarcoma), testis (seminoma, teratoma, embryonal carcinoma, teratocarcinoma, choriocarcinoma, sarcoma, interstitial cell carcinoma, fibroma, fibroadenoma, adenomatoid tumors, lipoma); Liver: hepatoma (hepatocellular carcinoma), cholangiocarcinoma, hepatoblastoma, angiosarcoma, hepatocellular adenoma, hemangioma, biliary passages; Bone: osteogenic sarcoma (osteosarcoma), fibrosarcoma, malignant fibrous histiocytoma, chondrosarcoma, Ewing's sarcoma, malignant lymphoma (reticulum cell sarcoma), multiple myeloma, malignant giant cell tumor chordoma,

osteochronfroma (osteocartilaginous exostoses), benign chondroma, chondroblastoma, chondromyxofibroma, osteoid osteoma and giant cell tumors; Nervous system: skull (osteoma, hemangioma, granuloma, xanthoma, osteitis deformans), head and neck cancer, meninges (meningioma, meningio sarcoma, gliomatosis), brain (astrocytoma, medulloblastoma, glioma, ependymoma, germinoma [pinealoma], glioblastoma multiform, oligodendroglioma, schwannoma, retinoblastoma, congenital tumors), spinal cord neurofibroma, meningioma, glioma, sarcoma); Gynecological: uterus (endometrial carcinoma), cervix (cervical carcinoma, pre-tumor cervical dysplasia), ovaries (ovarian carcinoma [serous cystadenocarcinoma, mucinous cystadenocarcinoma, unclassified carcinoma], granulosa-thecal cell tumors, Sertoll- Leydig cell tumors, dysgerminoma, malignant teratoma), vulva (squamous cell carcinoma, intraepithelial carcinoma, adenocarcinoma, fibrosarcoma, melanoma), vagina (clear cell carcinoma, squamous cell carcinoma, botryoid sarcoma (embryonal rhabdomyosarcoma), fallopian tubes (carcinoma), breast; Hematologic: blood (myeloid leukemia [acute and chronic], acute lymphoblastic leukemia, chronic lymphocytic leukemia, myeloproliferative diseases, multiple myeloma, myelodysplastic syndrome), Hodgkin's disease, non-Hodgkin's lymphoma [malignant lymphoma] hairy cell; lymphoid disorders; Skin: malignant melanoma, basal cell carcinoma, squamous cell carcinoma, Karposi's sarcoma, keratoacanthoma, moles dysplastic nevi, lipoma, angioma, dermatofibroma, keloids, psoriasis, Thyroid gland: papillary thyroid carcinoma, follicular thyroid carcinoma; medullary thyroid carcinoma, multiple endocrine neoplasia type 2A, multiple endocrine neoplasia type 2B, familial medullary thyroid cancer, pheochromocytoma, paraganglioma; and Adrenal glands: neuroblastoma.

In some embodiments, the subject has KRAS lung cancer. In some embodiments, the subject has triple negative breast cancer, HER2+ breast cancer, or luminal B breast cancer. The data presented herein demonstrates that each of KRAS LKB1+ lung cancer, triple negative breast cancer, HER2+ breast cancer, and luminal B breast cancer are consistently associated with upregulated STING levels. As such, in some embodiments, a subject having KRAS LKB1+ lung cancer, triple negative breast cancer, HER2+ breast cancer, or luminal B breast cancer is treated with one or more of (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; and (iii) an interferon gamma signaling agonist without measuring STING levels or the level of one or more SPARCs genes.

KRAS Lung Cancer

Non-small-cell lung cancer (NSCLC) is a heterogeneous disease, with multiple different oncogenic mutations. Approximately 25-30% of NSCLC patients present KRAS mutations, which confer poor prognosis and high risk of tumor recurrence. In the majority of cases, these KRAS mutations are missense mutations which introduce an amino acid substitution at position 12, 13, or 61 (e.g., an amino acid substitution at position 12, 13, or 61 of NCBI NP_004976.2). The result of these mutations is constitutive activation of KRAS signaling pathways. The role of KRAS mutations and their potential association with other common genetic lung cancer lesions (LKB1, P53) has recently been investigated in different cohorts of human lung adenocarcinomas using transcriptional, mutational, copy-number and proteomic data. These studies highlighted how LKB 1 inactivation is significantly associated with KRAS mutations compared to P53 deletion and that co-occurrence of KRAS mutation with inactivation of LKB 1 or P53 genes generates different tumor subsets with distinct biology, immune profiles, and therapeutic vulnerabilities. About half of NSCLCs with activating KRAS lesions also have deletions or inactivating mutations in the serine/threonine kinase 11 (LKB1) gene. Loss of LKB1 on a KRAS-mutant background may represent a significant source of heterogeneity contributing to poor response to therapy. LKB1 mutations associated with lung cancer are extensively characterized in the art. Exemplary LKB 1 mutations can be found, for example, in Kaufman et al, Cancer Research, 2016, the entirety of which is incorporated herein by reference.

The genotype of lung cancer can be determined by means readily known to those of skill in the art for assessing the genotype and/or levels of the markers, e.g., KRAS, LKB1, and p53, as described, for example, in Sholl, (Transl Lung Cancer Res. 2017 6(5): 560-569) including but not limited to, determining the genomic sequence of marker, e.g., by DNA sequencing or allele specific PCR, determining expression of the marker, e.g., by northern analysis, quantative PCR, or microarray analysis, or determining protein levels of the marker, e.g., by western analysis, mass spectrometry, immunohistochemostry, ect.

Breast Cancer

Breast cancers can be classified at the molecular level based on their genes and proteins by dividing them into four main molecular subtypes: HER2-positive, luminal A, luminal B and triple-negative.

One in five invasive breast cancers is HER2-positive, making this one of the more common breast cancer subtypes in the United States. HER2-positive cancers are ER- and PR- negative and human epidermal growth factor receptor 2 (HER2)-positive. HER2-positive breast cancer cells carry excess copies of the HER2 gene, which makes HER2-protein receptors, found on breast cells. Luminal A breast cancer is the most common subtype of breast cancer for every race and age. These tumors tend to be estrogen receptor (ER)-positive and progesterone receptor (PR)- positive and are typically slow growing.

Luminal B breast cancer includes tumors that are estrogen receptor positive,

progesterone receptor negative and HER2 or Ki67 positive. These tumors tend to grow more quickly than luminal A tumors.

In triple negative breast cancer (TNBC), the cells do not contain receptors for estrogen, progesterone or HER2. This type of breast cancer is usually invasive and usually begins in the breast ducts.

The classification of breast cancer can be determined by means readily known to those of skill in the art and is described, e.g., in Russnes et ah, (Am Jounal of Pathology 2017 187(10): 2152-2162) for assessing the genotype and/or levels of the markers, e.g., HER2, ER, PR, and Ki67 including but not limited to, determining the genomic sequence of marker, e.g., by DNA sequencing or allele specific PCR, determining expression of the marker, e.g., by northern analysis, quantative PCR, or microarray analysis, or determining protein levels of the marker, e.g., by western analysis, mass spectrometry, immunohistochemistry, ect.

The terms“administer,”“administering,” or“administration,” as used herein refers to implanting, absorbing, ingesting, injecting, or inhaling the one or more therapeutic agents.

As used herein, the term“treating” refers to the application or administration of a composition including one or more active agents to a subject, who has cancer, a symptom of cancer, or a predisposition toward the disease, with the purpose to cure, heal, alleviate, relieve, alter, remedy, ameliorate, improve, or affect the disorder, the symptom of the disease, or the predisposition toward the disease.

Alleviating cancer includes delaying the development or progression of the disease, or reducing disease severity. Alleviating the disease does not necessarily require curative results. As used therein, "delaying" the development of a disease (such as cancer) means to defer, hinder, slow, retard, stabilize, and/or postpone progression of the disease. This delay can be of varying lengths of time, depending on the history of the disease and/or individuals being treated. A method that "delays" or alleviates the development of a disease, or delays the onset of the disease, is a method that reduces probability of developing one or more symptoms of the disease in a given time frame and/or reduces extent of the symptoms in a given time frame, when compared to not using the method. Such comparisons are typically based on clinical studies, using a number of subjects sufficient to give a statistically significant result.

"Development" or "progression" of a disease means initial manifestations and/or ensuing progression of the disease. Development of the disease can be detectable and assessed using standard clinical techniques as well known in the art. However, development also refers to progression that may be undetectable. For purpose of this disclosure, development or progression refers to the biological course of the symptoms. "Development" includes occurrence, recurrence, and onset. As used herein "onset" or "occurrence" of cancer includes initial onset and/or recurrence.

An“effective amount” refers to an amount sufficient to elicit the desired biological response, i.e., treating the cancer. As will be appreciated by those of ordinary skill in this art, the effective amount of the compounds described herein may vary depending on such factors as the desired biological endpoint, the pharmacokinetics of the compound, the condition being treated, the mode of administration, and the age and health of the subject. An effective amount includes, but is not limited to, that amount necessary to slow, reduce, inhibit, ameliorate or reverse one or more symptoms associated with cancer. For example, in the treatment of cancer, such terms may refer to a reduction in the size of the tumor.

The compounds provided herein can be administered by any route, including enteral ( e.g ., oral), parenteral, intravenous, intramuscular, intra-arterial, intramedullary, intrathecal, subcutaneous, intraventricular, transdermal, interdermal, rectal, intravaginal, intraperitoneal, topical (as by powders, ointments, creams, and/or drops), mucosal, nasal, bucal, sublingual; by intratracheal instillation, bronchial instillation, and/or inhalation; and/or as an oral spray, nasal spray, and/or aerosol. Specifically contemplated routes are oral administration, intravenous administration (e.g., systemic intravenous injection), regional administration via blood and/or lymph supply, and/or direct administration to an affected site. In general, the most appropriate route of administration will depend upon a variety of factors including the nature of the agent (e.g., its stability in the environment of the gastrointestinal tract), and/or the condition of the subject (e.g., whether the subject is able to tolerate oral administration).

The exact amount of a compound required to achieve an effective amount will vary from subject to subject, depending, for example, on species, age, and general condition of a subject, severity of the side effects or disorder, identity of the particular compound, mode of

administration, and the like. The desired dosage can be delivered three times a day, two times a day, once a day, every other day, every third day, every week, every two weeks, every three weeks, or every four weeks. In certain embodiments, the desired dosage can be delivered using multiple administrations ( e.g ., two, three, four, five, six, seven, eight, nine, ten, eleven, twelve, thirteen, fourteen, or more administrations).

As used herein, the term“in combination” refers to the use of more than one therapeutic agent. The use of the term "in combination" does not restrict the order in which the therapeutic agents are administered to a subject with neoplasia. A first therapeutic agent, such as (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; (iii) an interferon gamma signaling agonist; (iv) a DNA methyltransferase inhibitor; or (v) an EZH2 inhibitor, can be administered prior to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks before), concomitantly with, or subsequent to (e.g., 5 minutes, 15 minutes, 30 minutes, 45 minutes, 1 hour, 2 hours, 4 hours, 6 hours, 12 hours, 24 hours, 48 hours, 72 hours, 96 hours, 1 week, 2 weeks, 3 weeks, 4 weeks, 5 weeks, 6 weeks, 8 weeks, or 12 weeks after) the administration of a second therapeutic agent, such as (i) a STING agonist; (ii) an Immune Checkpoint Blockade (ICB) agent; (iii) an interferon gamma signaling agonist; (iv) a DNA methyltransferase inhibitor; or (v) an EZH2 inhibitor described herein, to a subject. Thus, a first agent can be administered separately, sequentially or simultaneously with the second therapeutic agent.

In some embodiments, levels of STING and/or levels of one or more SPARCS genes are determined in a subject and a therapy is selected based on the level(s). In some embodiments, the therapy is a STING agonist. In some embodiments, the therapy is an Immune Checkpoint Blockade (ICB) agent. In some embodiments, the therapy is an interferon gamma signaling agonist. In some embodiments, the therapy is a DNA methyltransferase inhibitor. In some embodiments, the therapy is an EZH2 inhibitor.

In one embodiment, the STING agonist is a cyclic dinucleotide or a chemical molecule that binds to and activates STING, e.g., a cyclic dinucleotide comprising purine or pyrimidine nucleobases (e.g., adenosine, guanine, uracil, thymine, or cytosine nucleobases). In some embodiments, the nucleobases of the cyclic dinucleotide comprise the same nucleobase or different nucleobases. In some embodiments, the STING agonist comprises an adenosine or a guanosine nucleobase. In some embodiments, the STING agonist comprises one adenosine nucleobase and one guanosine nucleobase. In some embodiments, the STING agonist comprises two adenosine nucleobases or two guanosine nucleobases. In some embodiments, the STING agonist comprises a modified cyclic dinucleotide, e.g., comprising a modified nucleobase, a modified ribose, or a modified phosphate linkage. In some embodiments, the modified cyclic dinucleotide comprises a modified phosphate linkage, e.g., a thiophosphate. In some

embodiments, the STING agonist comprises a cyclic dinucleotide (e.g., a modified cyclic dinucleotide) with 2', 5' or 3', 5' phosphate linkages. In some embodiments, the STING agonist comprises a cyclic dinucleotide (e.g., a modified cyclic dinucleotide) with Rp or Sp

stereochemistry around the phosphate linkages.

The bacteria-produced cyclic dinucleotides, c-di-AMP, c-di-GMP and 3'3'-cGAMP, are agonists of STING. Other exemplary STING agonists include, e.g., cGAMP, c-di-GAMP, 2', 3'- cGAMP, 2’3’-c-di-AM(PS)2 (Rp,Rp), c-AIMP; (3\2')c-AIMP; (2',2')c-AIMP; (2\3')c-AIMP; c- AIMP(S); e-(dAMP-d!TMP); c-(dAMP-2'FdIMP); c-(2'FdAMP-2'FdIMP); (2 , ,3 , )c-(AMP-

2'FdIMP); c-PTdAMPfSl^'FdlMPTS)]; c-[2'FdAMP(S)-2 ' FdIMP(S)](POM)2; Rp,Rp dithio 2', 3' c-di-AMP (e.g., Rp,Rp-dithio c- [ A(2 ',5 ')p A(3 ',5 ')p] ) or a cyclic dinucleotide analog thereof; c-[G(2 ',5 ')pG(3 ',5 * )p] or a dithio ribose 0- substituted derivative thereof; c- [A(2',5')pA(3',5')p] or a dithio ribose 0-substituted derivative thereof, 2 '-O-propargyl-cyclic-

[A(2',5')pA(3',5')p] (2'-0-propargyl . ML-CDA), CL614 (Invitrogen), CL614 (Invitrogen),

CL656 (Invitrogen), 2’3’-cGAM(PS)2 (Rp/Sp), cyclic [FdG(3 ,5’ )pFd A(3’ ,5’ )p] , cyclic

[FdA(3’ ,5’ )pFd A(3 \5’ )p] , 2’3’-c-di-AMP, cyclic [FdG(3\5’)pFdG(3\5’)p], 2’3’-c-di-GMP, c- di-EVlP, 5601 (Tocris), G10 (Tocris), SB 11285 (Spring Bank Pharmaceuticals), MK-1454 (Merck) , or ADU-S100 (Aduro Biotech). In another embodiment, the unnatural or synthetic dinucleotide 2'2'-cGAMP is an agonist of STING In some embodiments, the STING agonist can comprise a flavonoid. In other embodiments, the STING agonist can consist of a flavonoid. Suitable flavonoids include, but are not limited to, lO-(carboxymethyl)-9(lOH)acridone (CMA), 5,6-Dimethylxanthenone-4-acetic acid (DMXAA), methoxyvone, 6,4'-dimethoxyflavone, 4'- methoxyflavone, 3',6'-dihydroxyflavone, 7,2'-dihydroxyflavone, daidzein, formononetin, retusin 7-methyl ether, xanthone, or any combination thereof. In some embodiments, the STING agonist is a dimeric amidobenzimidazole. In some embodiments, the STING agonist is SRCB- 0074 (See Chin et ah, Open 20l8;3). Other exemplary STING agonists are disclosed, e.g., in PCT Publication Nos. WO 2014/189805 and WO 2014/189806, and U.S. Publication Nos. 2015/0056225 2016/0287623 and 2018/0028553, each of which is incorporated by reference.

In some embodiments, a STING agonist comprises the STING protein or a fragment thereof. In some embodiments, a fragment of the STING protein comprises 10 or more consecutive amino acid residues of SEQ ID NO: 1, e.g., 15 or more, 20 or more, 25 or more, 30 or more, 35 or more, 50 or more, 100 or more, or 200 or more consecutive amino acid residues of SEQ ID NO: 1. In some embodiments, the STING agonist is a protein that is about 80%, 85%, 90%, 95%, or 99% identical to SEQ ID NO: 1.

In some embodiments, a STING agonist comprises an mRNA, e.g., a synthetic mRNA expressing the STING protein or a fragment thereof. As used herein the term,

“synthetic mRNA” refers to a mRNA produced through an in vitro transcription reaction or through artificial (non-natural) chemical synthesis or through a combination thereof. In some embodiments, the synthetic RNA further comprises a poly A tail, a Kozak sequence, a 3' untranslated region, a 5' untranslated region, or any combination thereof. Poly A tails in particular can be added to a synthetic RNA using a variety of art-recognized techniques, e.g., using poly A polymerase (Yokoe, et al. Nature Biotechnology. 1996; 14:1252-1256), using transcription directly from PCR products, or by ligating to the 3' end of a synthetic RNA with RNA ligase (see, e.g., Molecular Cloning A Laboratory Manual, 2 nd Ed., ed. by Sambrook, Fritsch and Maniatis (Cold Spring Harbor Laboratory Press: 1991 edition)).

In some embodiments, a STING agonist comprises a vector encoding the STING protein or a fragment thereof. Viral vectors are engineered to transduce one or more desired nucleic acids into a cell. In some embodiments, the viral vector is a retroviral transfer vector, an adenoviral vector, a lentiviral vector or an adeno-associated viral vector. In some embodiments, the viral vector is an adenoviral vector, and the adenoviral vector is a subgroup A, subgroup B, subgroup C, subgroup D, subgroup E, or subgroup F adenoviral vector. In some embodiments, the viral vector is a lentiviral vector, and the lentiviral vector is an HIV, SIV, FIV, EIAVor ovine lentiviral vector. In some embodiments, the viral vector is an adeno-associated viral vector, and the adeno-associated viral vector is an AAV1, AAV2, AAV5, AAV6, AAV6.2, AAV7, AAV8, AAV9, AAV 10 or AAV 11 adeno-associated viral vector. In some

embodiments, the viral vector is a chimeric viral vector. In one embodiment of any one of the methods provided herein, the chimeric viral vector is an AAV-adenoviral transfer vector.

In any of the foregoing aspects and following embodiments, an Immune Checkpoint Blockade (ICB) agent is, for example, an antagonist of surface proteins which are members of either the TNF receptor or B7 superfamilies, including without limitation, programmed death 1 (PD-l), programmed death ligand 1 (PD-L1), cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), V-domain Ig suppressor of T cell activation (VISTA), programmed death ligand 2 (PD-L2), indoleamine 2,3-dioxygenase (IDO), arginase, B7 family inhibitory ligand B7-H3, B7 family inhibitory ligand B7-H4, lymphocyte activation gene 3 (LAG3), 2B4, B and T

lymphocyte attenuator (BTLA), T cell membrane protein 3 (TIM3; also known as HAVcr2), adenosine A2a receptor (A2aR), a killer inhibitory receptor, CD160, CD244, TIGIT, BMA, and/or signal transducer and activator of transcription (STAT)3. In some embodiments, the antagonist of the is agent that binds to and antagonizes the surface protein, e.g., PD-l, PD-L1, CTLA-4, VISTA, PD-L2, IDO, B7-H3, B7-H4, LAG3, 2B4, BTLA, TIM3, A2aR, CD 160, CD244, TIGIT, BMA, and/or STAT3 is (i) an antisense molecule directed against a nucleic acid encoding the surface protein, (ii) an adnectin directed against a nucleic acid encoding the surface protein, (iii) a single stranded or double stranded RNAi inhibitor of the surface protein, (iv) a small molecule inhibitor of the surface protein, or (v) an antibody that specifically binds the surface protein.

In any of the foregoing aspects and embodiments, the PD-l antagonist is, for example, an agent that binds to and antagonizes PD-l. Such agents can be, for example, a peptide that binds PD-l. Such agents can be a humanized antibody that selectively binds PD-l. In some

embodiments, the humanized antibody that selectively binds PD-l is lambrolizumab, nivolumab, pembrolizumab, pidilizumab, MED 1-0680, REGN2810, or AMP-224. In some embodiments, the humanized antibody that selectively binds PD-l is nivolumab, pembrolizumab, or pidilizumab.

In some embodiments, the antagonist is (i) an antisense molecule directed against PD-l nucleic acid, (ii) an adnectin directed against PD- 1 nucleic acid, (iii) a single stranded or double stranded RNAi inhibitor of PD-l, and/or (iv) a small molecule inhibitor of PD-l.

In any of the foregoing aspects and embodiments, the PD-L1 antagonist is, for example, an agent that binds to and antagonizes PD-L1. Such agents can be, for example, a peptide that binds PD-L1. Such agents can be a humanized antibody that selectively binds PD-L1. In some embodiments, the humanized antibody that selectively binds PD-L1 is BMS-936559/ MDX- 1105, MPDL33280A/ RG7446/ atezolizumab, MSB0010718C/ avelumab, MIH1,

pembrolizumab, or MEDI4736/ durvalumab. In some embodiments, the antagonist is (i) an antisense molecule directed against PD-L1, (ii) an adnectin directed against PD-L1, (iii) a single stranded or double stranded RNAi inhibitor of PD-L1, or (iv) a small molecule inhibitor of PD- Ll.

B7-H3 checkpoint inhibitors, include, without limitation, antibodies neutralizing human B7-H3 (e.g. MGA271 disclosed as BRCA84D and derivatives in U.S. Patent Application Publication No. 2012/0294796, incorporated herein by reference). In some embodiments, the antagonist is (i) an antisense molecule directed against B7-H3, (ii) an adnectin directed against B7-H3, (iii) a single stranded or double stranded RNAi inhibitor of B7-H3, or (iv) a small molecule inhibitor of B7-H3.

B7H4 checkpoint inhibitors include, without limitation, antibodies to human B7H4 (disclosed in WO 2013025779 Al, and in U.S. Patent Application Publication No.

2014/0294861, incorporated herein by reference) or soluble recombinant forms of B7H4 (such as disclosed in U.S. Patent Application Publication No. 2012/0177645, incorporated herein by reference, or Anti-human B7H4 clone H74: eBiocience #14-5948). In some embodiments, the antagonist is (i) an antisense molecule directed against B7-H4, (ii) an adnectin directed against B7-H4, (iii) a single stranded or double stranded RNAi inhibitor of B7-H4, or (iv) a small molecule inhibitor of B7-H4.

TIM3 checkpoint inhibitors include, without limitation, antibodies targeting human TIM3 (e.g. as disclosed in U.S. Pat. No. 8,841,418, incorporated herein by reference, or the anti human TIM3, blocking antibody F38-2E2 disclosed by Jones et ah, J Exp Med., 205(l2):2763- 79 (2008)). In some embodiments, the antagonist is (i) an antisense molecule directed against TIM3, (ii) an adnectin directed against TIM3, (iii) a single stranded or double stranded RNAi inhibitor of TIM3, or (iv) a small molecule inhibitor of TIM3.

TIGIT checkpoint inhibitors preferably inhibit interaction of TIGIT with polovirus receptor (CD155) and include, without limitation, antibodies targeting human TIGIT, such as those disclosed in U.S. Pat. No. 9,499,596 and U.S. Patent Application Publication Nos.

20160355589, 20160176963 and polovirus variants such as those disclosed in U.S. Pat. No. 9,327,014. In some embodiments, the antagonist is (i) an antisense molecule directed against TIGIT, (ii) an adnectin directed against TIGIT, (iii) a single stranded or double stranded RNAi inhibitor of TIGIT, or (iv) a small molecule inhibitor of TIGIT.

In any of the foregoing aspects and embodiments, the CTLA-4 antagonist is, for example, an agent that binds to and antagonizes CTLA-4. Such agents can be, for example, a peptide that binds CTLA-4. Such agents can be a humanized antibody that selectively binds CTLA-4. In some embodiments, the humanized antibody that selectively binds CTLA-4 is ipilimumab or tremelimumab. In some embodiments, the CTLA-4 antagonist is (i) an antisense molecule directed against CD80, CD86, and/or CTLA-4 nucleic acid, (ii) an adnectin directed against CD80, CD86, and/or CTLA-4 nucleic acid, (iii) a single stranded or double stranded RNAi inhibitor of CD80, CD86, and/or CTLA-4, or (iv) a small molecule inhibitor of CD80, CD86, or CTLA-4.

In any of the foregoing aspects and embodiments, the VISTA antagonist is, for example, an agent that binds to and antagonizes VISTA. Such agents can be, for example, a peptide. Such agents can be an inhibitory antibody directed to VISTA. In some embodiments, the agent that binds to and antagonizes VISTA is a humanized antibody. In some embodiments, the agent that binds to and antagonizes VISTA is (i) an antisense molecule directed against VISTA nucleic acid, (ii) an adnectin directed against VISTA nucleic acid, (iii) a single stranded or double stranded RNAi inhibitor of VISTA, or (iv) a small molecule inhibitor of VISTA.

Exemplary interferon gamma signaling agonists include, e.g., interferon gamma or interferon gamma variants. In some embodiments, an interferon gamma or interferon gamma variant comprises Actimmune (Genentech). In some embodiments, an interferon gamma or interferon gamma variant comprises recombinant interferon gamma described, for example, in Gray et al., Nature 295, 503-508 (1982); U.S. Pat. Nos. 4,762,791, 4,929,544, 4,727,138, 4,925,793, 4,855,238, 5,582,824, 5,096,705, 5,574,137, and 5,595,888, the contents of each of which are incorporated by reference herein in their entirety. In some embodiments, an interferon gamma or interferon gamma variant comprises recombinant interferon gamma variants described in 7,038,015 or 6,958,388, the contents of each of which are incorporated by reference herein in their entirety.

Interferon gamma has the following amino acid sequence:

mkytsyilaf qlcivlgslg cycqdpyvke aenlkkyfna ghsdvadngt lflgilknwkeesdrkimqs qivsfyfklf knfkddqsiq ksvetikedm nvkffnsnkk krddfekltnysvtdlnvqr kaiheliqvm aelspaaktg krkrsqmlfr grrasq (SEQ ID NO: 110)

In some embodiments, an interferon gamma signaling agonist comprises the interferon gamma protein or a fragment thereof. In some embodiments, a fragment of the interferon gamma protein comprises 10 or more consecutive amino acid residues of SEQ ID NO: 110, e.g., 15 or more, 20 or more, 25 or more, 30 or more, 35 or more, 50 or more, 100 or more, or 200 or more consecutive amino acid residues of SEQ ID NO: 110. In some embodiments, the interferon gamma signaling agonist is a protein that is about 80%, 85%, 90%, 95%, or 99% identical to SEQ ID NO: 110. In some embodiments, an interferon gamma signaling agonist comprises an mRNA, e.g., a synthetic mRNA expressing the interferon gamma protein or a fragment thereof. In some embodiments, an interferon gamma signaling agonist comprises a retroviral vector encoding the interferon gamma protein or a fragment thereof.

In some embodiments, the interferon gamma signaling agonist comprises baicalin, poly I:C, IL-18, IL-2, IL-12, IL-27, an IL-27 hyperkine, IRF-10, or PTPN2 inhibitors

In some embodiments, the interferon gamma signaling agonist comprises mitogens. Exemplary mitogens include but are not limited to lectins, phytohemagglutinin, concanavalin A, pokeweed mitogen, lipopolysaccharide, endotoxin, lipid A, polysaccharide and bacterium.

In some embodiments, the interferon gamma signaling agonist comprises a nucleic acid. In some embodiments, the nucleic acid comprises bacterial DNA. In some embodiments, the nucleic acid comprises an oligonucleotide containing CpG motifs.

In some embodiments, the interferon gamma signaling agonist comprises a small molecule mimic of interferon gamma. Exemplary small molecule mimics of interferon gamma are described, for example, in US 10197557, the entire contents of which is incorporated herein by reference in its entirety.

Exemplary DNA methyltransferase inhibitors include, e.g., azacitidine, decitabine, zebularine, NPEOC-DAC, CP-4200, RX-3117, cytosine analogues, thio-cytidine derivatives, e.g., T-dCyd and 5-aza-T-dCyd, decitabine-p-deoxyguanosine (SGI-110), SAM analogues, SAH analogues, SGI- 1027, alcyne derivatives, cyclopenta derivatives, cyclohexathiophene derivatives, tryptophane derivates, e.g., RG108, procainamide derivatives, flavonoid derivatives, curcumin, psammaplin, hydralazine, disulfiram, 5-fluro-2’-deoxycitidine, 5-azacytidine, 5-aza- 2'-deoxycytidine, 5,6-dihydro-5-azacytidine, 5-fluoro-2'-deoxycytidine, l-(beta-D- ribofuranosyl)- 1 ,2-dihydropyrimidin-2-one, 5-aza-2'-deoxycytidine-p-deoxyguanosine, fluorocyclopentenylcytosine, 2-(p-nitrophenyl) ethoxycarbonyl-5-aza-2'-deoxycytidine, 4'-thio- 2'-deoxycytidine, 5-aza-4'-thio-2'-deoxycytidine, l-beta-D-arabinofuranosyl-5-azacytosine, hydralazine, procaine, mithramycin A, nanaomycin A, N-(4-((2-amino-6-methylpyrimidin-4- yl)amino)phenyl)-4-(quinolin-4-ylamino)benzamide, N -phthalyl-L-tryptophan, N -phthalyl-L tryptophan derivatives, alkine derivatives, halomon, S-adenosyl-L-methionine analogues, S- adenosyl-L-homocysteine, S-adenosyl-L-homocysteine analogues, MG98, miR-29a, miR-29c, Sinefungin, procainamide, procainamide derivatives, procainamide-N -phthalyl-L-tryptophan conjugates, cyclopentathiophene derivatives, cyclohexathiophene derivatives, flavone derivatives, 3-nitroflavone derivatives, flavanones derivatives, 3-chloro-3-nitroflavanone derivatives, diclone, laccaic acid, acridine, 5,5'-Methylenedisalicylic acid, 4-(2-((5-Chloro-2- methoxybenzoyl)amino)ethyl)hydrocinnamic acid, 4-Chloro-N-(4-hydroxy- l-naphthalenyl)-3- nitro-benzenesulfonamide, (S)-3-(lH-Indol-3-yl)-2-(5-nitro-l,3-dioxo-l,3-dihydro-isoin dol-2- yl)-propionic acid, (R)-2-(l,3-Dioxo-5-phenylethynyl-l,3-dihydro-isoindol-2-yl)- 3-(lH-indol-3- yl)-propionic acid, (S)-2-(2,6-Dioxo-piperidin-l-yl)-3-(lH-indol-3-yl)-propionic acid,

N-hydroxy-4-(2-(4-aminobenzamido)-ethylcarbamoyl)butanami de, 4-amino-N-(2-(ethyl(3- (hydroxyamino)-3-oxopropyl)amino)ethyl)benzamide, N<l> -(2-(4-aminobenzamido)ethyl)- N<l> -ethyl-N<6> -hydroxyadipamide, tetraethylthiuramdisulfide,

(-)-epigallocatechin-3-gallate, genistein, psammaplin A, psammaplin derivatives, and anti-DNA methyltransferase antibodies.

Non-limiting examples of EZH2 inhibitors include S-adenosyl-methionine-competitive small molecule inhibitors. In particular non-limiting embodiments, the EZH2 inhibitor is derived from tetramethylpiperidinyl compounds. Further non-limiting examples include

UNC1999, 3 -Deazaneplanocin A (DZNcp), Ell, EPZ-5676, EPZ-6438, GSK343, EPZ005687, EPZO 11989, GSK126, CAS #1346574-57-9, (S)-l-(sec-butyl)-N-((4,6-dimethyl-2-oxo-l,2- dihydropyridin-3-yl)methyl)-3-methyl-6-(6-(piperazin-l-yl)py ridin-3-yl)-lH-indole-4- carboxamide, DZNep, tazemetostat, anti-EZH2 antibodies, and siRNA directed against EZH2.

Example 1: Tumor innate immunity primed by specific interferon-stimulated endogenous retroviruses

Mesenchymal tumor subclones secrete pro-tumorigenic cytokines and promote treatment resistance, but the underlying mechanism remains poorly understood. Here an epigenetically regulated subclass of endogenous retroviruses (ERVs) that engages innate immune signaling in mesenchymal cells is identified. Stimulated 3 Prime Antisense Retroviral Coding Sequences (SPARCS) are oriented inversely in 3’ETTRs of certain interferon-inducible genes silenced by EZH2. De-repression of these loci results in dsRNA generation following IFNy exposure due to bi-directional transcription from the STAT1 -activated gene promoter and the 5’ LTR of the antisense ERV. dsRNA sensing by MAVS fuels activation of TBK1, IRF3, and STAT1 signaling, sustaining a positive feedback loop. SPARCS induction in human tumors is tightly associated with B2M and MHC class 1 antigen expression, mesenchymal markers, and downregulation of chromatin modifying enzymes, including EZH2. Analysis of cell lines poised to induce high levels of SPARCS expression reveals strong association with an AXL positive mesenchymal cell state. While SPARCS high tumors are marked by immune infiltration, they also exhibit multiple features of immune suppression including checkpoint gene expression and myeloid cell infiltration. Together, these data unveil a novel subclass of ERVs whose de repression triggers pathologic innate immune signaling in cancer, with important implications for cancer immunotherapy.

Subclonal tumor heterogeneity promotes cancer progression via different growth and metastatic properties, drug sensitivity, and paracrine signaling 1 4 . Resistant small cell lung cancer (SCLC), for example, undergoes a mesenchymal state switch induced by RAS/MET signaling or chemotherapy (e.g. H69M or H69AR cells) (Figure 5A) 5,6 . A mesenchymal AXL/NF-kB high state also promotes therapeutic resistance 7 10 , though remains incompletely defined.

Enhanced innate immune and RAS signaling are noted in H69M cells, including elevated phosphorylated-TBKl (pTBKl), pIRF3, IKKe and NF-kB gene sets (Figures 1A and 5B) and multiple secreted cytokines/chemokines (Figures 1B and 5C). pTBKl levels were preferentially increased in additional mesenchymal SCFC cell lines (Figures 5D and 5E), and subclonally in human and murine Rb^/pSS 1 ^ derived SCFC tumors (Figures 1C and 5F-H). H69 mesenchymal cell lines also uniquely attracted T cells and monocytes (Figures 6A-E), prompting the exploration of immune checkpoint activation. Interestingly, H69M contained a PD-Fl hlgh , CD44 hlgh fibroblastic subclone (Figures 1D, 7A, and 7B) that was the source of elevated pTBKl and cytokine/chemokine production (Figures 1E and 1F).

H69M PD-Fl hlgh cells reverted morphologically over time and were genomically similar to parental H69 cells (Figures 7C and 7D). These findings suggested an epigenetic mechanism of innate immune activation. Since endogenous retroviruses (ERVs) undergo epigenetic silencing, they were assessed for potential de -repression of a recently described ERV panel 11,12 . H69M-PD-Fl hlgh cells exhibited marked upregulation of MLT1C49 (Figure 1G), an ERV poised to generate dsRNA due to antisense orientation in the 3’FTTR of TRIM22, an IFN-stimulated gene (Figure 1H) 13 . Because of this unique feature, all 3’FTTR repeat elements from Ref Seq were intersected with H69M upregulated genes 5 identifying TRIM22 and 14 other genes with 3’ UTR antisense ERVs, including TRIM38 which contained two ( MLT1J , MLT1A) (Figure II). Given their potential to generate dsRNA and spark innate immune signaling these ERVs are termed Stimulated 3 Prime Antisense Retroviral Coding Sequences (SPARCS). Since ERV dsRNAs are sensed by MAVS or reverse-transcribed and detected via the cGAS-cGAMP STING pathway 14 , and since STING especially was upregulated in concert with this phenotype (Figure 7E), CRISPR was used to delete MAVS and/or STING in H69M cells. MAVS or combined MAVS/ STING deletion in H69M cells strongly impaired TBK1 and IRF3 phosphorylation (Figure 1J), decreased multiple cytokines/chemokines, including CXCF10, CCF5, and CCF2 (Figures 1K and 1F) and significantly suppressed T cell and monocyte attraction (Figures 7F and 7G). Deletion of MAVS and/or STING also reverted the mesenchymal phenotype (Figure 7H), increasing E-cadherin and decreasing Vimentin gene and protein expression (Figures 1J and 71). Thus ERV sensing of SPARCS directly contributes to this cellular state.

In contrast to H69M-PD-Fl hlgh cells, which secreted IFNy at baseline (Figure 1F), chemoresistant H69AR cells required exogenous IFNy for robust PD-F1 induction (Figure 2A) and lacked basal MLT1C49 expression (Figure 8A). SPARCS inducibility were therefore explored by transient IFNy pulse treatment of H69AR compared with H69 cells. Even a 10 minute IFNy pulse resulted in marked induction of TRIM22 and MLT1C49 in H69AR cells, correlating with PD-L1 mRNA and secretion of multiple cytokines, including CXCF10 and CCF2 (Figures 2B, 2C, 8B and 8C). Indeed, H69AR cells showed gain of chromatin accessibility around TRIM22, TRIM38, and multiple other SPARCS loci, but not PD-LJ CXCL10 or CCL2 (Figures 2D and 8D). Because H69M and H69AR cells downregulated EZH2 15 (Figure 2E), it was next tested whether EZH2 is involved in silencing SPARCS. EZH2 inhibitor treatment of H69 cells over 6 days enhanced IFNy induced cytokine secretion (Figure 2F), similar to IFNy pulsed H69AR cells (Figure 8C), and de-repressed the same SPARCS as in H69AR cells (R 2 = 0.87488) (Figures 2G, 9A and 9B). Thus, SPARCS loci are normally silenced and protected from IFN exposure by EZH2, but de-repressed in mesenchymal SCFC subclones.

It was sought to determine if SPARCS promote positive feedback signal amplification, which would explain their propensity to be silenced. High levels of dsRNA preferentially produced from MLT1C49, MLT1J and MLT1A were detected in 10 minute IFNy pulsed H69AR cells, even after 24 hours (Figure 2H). TAG-aided sense/antisense transcript detection (TASA- TD) PCR 16 confirmed that dsRNA resulted from bidirectional transcription of MLT1C49, MLT1J, and MLT1A (Figure 9C). IFNy pulse treatment uniquely hyperactivated and sustained pTBKl and pIRF3 in addition to pSTATl in H69AR but not H69 cells (Figure 21), which further amplified SPARCS expression, pTBKl, and its effector cytokines over time (Figures 2J, 9D and 9E). Induction of type I/I I IFNs was also confirmed by transfection of the dsRNA mimic Poly(LC) in H69AR cells (Figure 9F) and SPARCS upregulation by direct IFNa/b exposure or by Poly(LC) itself (Figures 9F and 9G), consistent with positive feedback induced by dsRNA and type I IFN. Furthermore, exposure of untreated H69AR cells to IFNy primed conditioned medium also activated TBK1 and STAT1 and induced CXCL10, IRNb and ERV expression (Figures 10A-D). As expected, treatment with the JAK1/2 inhibitor Ruxolitinib disrupted this circuit, whereas TBKI/IKKk inhibition with MRT67307, partially inhibited downstream CXCL10 expression (Figures 10E-G).

The contribution of SPARCS ERV sensing to feedforward signaling in H69AR cells was also directly assessed. As expected, MAVS deletion impaired Poly(I:C)-induced CXCL10 and type I/II IFN expression and CXCL10 secretion (Figures 2K, 10H, and 101). MAVS and especially MAVS I STING knockout (KO) H69AR cells downregulated multiple cytokines/chemokines following low dose IFNy pulse treatment including CXCL10 as well as VEGF (Figures 2L and 2M). Tumorigenicity of sgMAVS-H69AR cells in nude mice was thus also significantly impaired relative to control (Figure 2N).

To determine the broader relevance of SPARCS and to begin to explore the relationship to immune contexture, next the expression of the 15 gene SPARCS signature across the Cancer Genome Atlas (TCGA) (Pancanl2, n=3602 tumors) 17 was ranked using single sample gene set enrichment analysis (ssGSEA) 18 (Figure 3A). Top gene sets co-enriched with SPARCS (p<0.0l, FDR O.Ol) included epigenetic, TNF/NF-kB, inflammation/innate immunity, and RTK/KRAS signaling (Figure 3A), also observed in the Cancer Cell Line Encyclopedia (CCLE) (Figure 11A and). In contrast, none of these signatures correlated with 3’ UTR antisense ERVs from H69M downregulated genes (Figure 11B). The SPARCS high state also co-associated with mutations on chromosome 3p, including PBRM1 and SETD2, as well as oncogenic KRAS in CCLE (Figures 12A and B).

At the gene level, SPARCS expression correlated with markers of T cell ( CD4 , PR FI and GZMA) and myeloid ( CD40 , CD86, CD14, CD68) infiltration uniquely in TCGA. In both CCLE and TCGA datasets SPARCS associated with expression of MHC, APOBEC, immunosuppressive factors such as CD274 (PD-L1), PDCD1LG2 (PD-L2), C10orf54 (VISTA), TIM3, and IDOl, and EMT genes (Figures 12C and 12D). Expression of EZH2, DNMT3A, SETD2 and multiple SWI/SNF genes was inversely correlated with SPARCS in both datasets (Figures 12C and 12D). The top 1000 genes from TCGA and CCLE were next intersected, to isolate robust cancer cell autonomous genes co-regulated with SPARCS (Figure 3B). This identified B2M as the top ranked gene, followed by multiple MHC class 1 ( HLA-A , B, C, E, F) and class 1 related buytyrophilin (BTN) genes 19 , cytosolic RNA sensors ( IFIH1/MDA5 , DDX60 ) and the antigen processing machinery ( PSMB9 , TAPI , TAP2), all markers of a virally infected state (Figure 3B). TGFB1 and AXF were also top hits in this analysis (Figure 3B).

High SPARCS expression in TCGA was enriched in distinct cancer histologies beyond SCLC, including clear cell renal (KIRC), lung adenocarcinoma (LUAD), head/neck squamous (HNSC), and glioblastoma (GBM) (Figure 3C). SPARCS hlgh CCLE lines also included triple negative breast cancer (TNBC), and exhibited high relative expression of AXF, MET, VIM, TGFB1 and CD44, and low EZH2, SETD2 and SWI/SNF component expression (Figure 13A). To validate these findings, multiple cell lines were cultured, confirming that SPARCS hlgh cells exhibited mesenchymal morphology and elevated AXL, MET and Vimentin relative to SPARCS low cells (Figures 3D and 13B). Similar to the H69 model, SPARCS hlgh cells also exhibited increased STING levels, whereas MAVS was more evenly expressed (Figures 3D and 13A). It was further confirmed that IFNy pulse treatment of SPARCS hlgh cells significantly induced MFT1C49, CXCF10 and PD-F1 expression relative to SPARCS low cells, and inducible surface PD-L1 correlated with their high baseline CD44 expression (Figures 3E and 14A).

Immune cell GSEA 20 was next used to assess whether certain immune infiltrates might be associated with elevated SPARCS expression in TCGA (Figure 4A). Despite markers of cytotoxic T cells and an adaptive immune response (AIR), the top associated signatures were innate immune response (HR) and myeloid derived suppressor cells (MDSC), followed by neutrophils and macrophages (Figure 4A). Grouping of TCGA tumors into discrete SPARCS high and low categories (Figure 14B), confirmed robust and significant association of SPARCS high tumors with HR (q value = 9.68E-275), MDSC (q value = 7.73E-257), AIR (q value = 1.41E-201), neutrophils (q value = 2.41E-131) and macrophages (q value = 4.60E-126) (Figures 4B and 4C). Consistent with T cell and myeloid cell chemotaxis, CXCF10 and CCF2 gene expression in primary tumors was tightly associated with the SPARCS hlgh state (Figure 4D). Thus, myeloid cell infiltration may further contribute to an immunosuppressed microenvironment in SPARCS hlgh tumors.

Finally, ex vivo culture of patient-derived organotypic tumor spheroids (PDOTS) was utilized, which retain autologous tumor infiltrating lymphocytes 21 , to determine the potential translational relevance of these findings. Using multiplexed immunofluorescence, T cell infiltration of two different KRAS mutant non-small lung cancer specimens was confirmed (Figure 4E), one of which had SPARCS hlgh features including SETD2 inactivation and an APOBEC mutation pattern (NSCLC-l), in contrast to the other which exhibited STK11/TP53 co-mutation (NSCLC-2) and T cell localization between tumor nests. IFNy or Poly (I:C) treatment of NSCLC-l PDOTS markedly enhanced production of multiple cytokines/chemokines, especially CXCL10, and sensitized them to ex vivo PD-l blockade with nivolumab, in contrast to NSCLC-2 (Figures 4F-H and 14C-E). Thus hyperactivated IFN signaling associated with the SPARCS hlgh state can be directly ascertained from patient samples and may promote sensitivity to PD-l blockade.

Here SPARCS was identified as a novel subclass of ERVs silenced by EZH2 and poised to undergo positive feedback signal amplification due to their antisense localization in 3’ UTRs of IFN stimulated genes. Whereas prior reports utilized DNMT inhibition to uncover ERVs more generally 11,12 , the data reveal that mesenchymal tumor subclones with high AXL expression and low EZH2 levels trigger SPARCS expression when exposed to IFNy. This SPARCS high state was also associated with downregulation of multiple SWI/SNF components and RCC, potentially contributing to the immunogenicity recently reported following PBRM1 inactivation 22,23 . While SPARCS expression promotes MHC class 1 upregulation and T cell infiltration, activation of immune checkpoints and myeloid infiltration may promote tumor immune suppression, similar to a chronic virally infected state. Therapeutically, this may have important implications for drug combinations with PD-l blockade. For example, in contrast to potential antagonism by JAK1/2 inhibitors, therapies that activate JAK signaling such as PTPN2 inhibition 24 , inhibit certain chromatin regulators 22,23,25 , or target TBK1 21,24 , could alter SPARCS physiology to favor response.

Methods

Patients samples

SCLC and NSCLC human tumor samples were collected and analyzed according to Dana-Farber/Harvard Cancer Center IRB-approved protocols. These studies were conducted according to the Declaration of Helsinki and approved by Dana-Farber and Brigham and Women’s Hospital IRBs. Cell lines

The human SCLC cell lines H69, H69M, H69AR, H841, SHP77, H187, H345 and H524 were obtained from the laboratory of Dr. Joan Albanell and were authenticated following Short Tandem Repeat (STR) genotyping. Clear cell renal carcinoma (ccRCC) cell lines A704, A498, 786-0, 769-P and Caki-2 were obtained from the laboratory of Dr. William G. Kaelin Jr. and were authenticated following STR genotyping. HCC44 cell line was obtained from Broad Institute and was authenticated following STR genotyping. Jr. Jurkat T cells, THP-l monocytes, H196, MDA-MB-231, HCC1143, MDA-MB-468, H522, T47D, MDA-MB-453, H1436 and H2081 were obtained from the American Type Culture Collection (ATCC) (Rockville, MD) and used for all experiments before reaching 10 passages.

H69, H69M, H69AR, H841, SHP77, H187, H345, H524, H196, HCC44, MDA-MB-231, MDA-MB-453, MDA-MB-468, T47D, HCC1143, H522, H1436, H2081, THP-l, Jurkat and 769-P were cultured in RPMI-1640 (Thermo Fisher Scientific, #11875-119) containing 10% FBS (Gemini Bio-products, #100-106) and IX pen-strep (Gemini Bio-products, #400-109). 786- O and A498 were maintained in Dulbecco’s Modified Eagles Medium (DMEM) (Thermo Fisher Scientific, #11965-118) containing 10% FBS and IX pen-strep. Caki-2 cells were maintained in McCoy’s 5A medium (Life Technologies #16600108) supplemented with 10% FBS and IX Penicillin-Streptomycin. A704 cell lines was maintained in Eagle’s Minimum Essential Medium (EMEM) (Sigma, #M4780) supplemented with 2mM Glutamine (Life Technologies, #25030081), 1% Non-Essential Amino Acids (NEAA) (Life Technologies, #11140-050), lmM Sodium pyruvate (Life Technologies, #11360-070), 15% FBS and IX Penicillin- Streptomycin.

Compounds and treatments

Recombinant human IFN-g (#285-IF) IFN-a (#11100-1) and IFN-b (#8499-IF) proteins were purchased from R&D Systems (Minneapolis, MN) and reconstituted in sterile, deionized water. MRT67307 and Ruxolitinib were synthesized and purchased from Shanghai Haoyuan Chemexpress Co. Both drugs were reconstituted at 10 mM in DMSO and stored at -20°C. GSK126 (#S706l) was purchased from Selleck chemicals (Houston, TX) and reconstituted at 5 mM in DMSO and stored at -20°C.

For IFN pulse experiments, cells were pulsed 10 minutes with IFN-g 200 ng/mL or 10 ng/mL), IFN-a 10 000 U/mL), or IFN-b 10 000 U/mL), extensively washed, and chased in fresh media for an additional 24, 48 or 72 hours. To test drug effects on gene expression or protein secretion, IFN-g pulsed H69AR cells were treated with DMSO, ImM MRT67307 or 100 nM Ruxolitinib for 24, 48 and 72 hours.

For EZH2 inhibition experiments, H69 cells were treated with 5 mM GSK126 for 6 days. Drug was replenished every 3 days with both suspension and adherent cells carried each time. After the GSK126 treatment period, equal numbers of DMSO-treated and GSKl26-treated cells were exposed to either H 2 0 or 200 ng/mL IFN-g for 24 hours before harvesting of RNA or conditioned media (CM).

SCLC GEM model

All mouse experiments were conducted in accord with a Dana-Farber Cancer Institute Institutional Animal Care and Use Committee (IACUC) approved protocol. Primary tumor and metastasis tissue sections used in this study were from the genetically engineered mouse (GEM) model of SCLC consisting of the Rb^/pSS^ allelic genotype 26 . A total number of 24 slices of 1 mm thickness were collected providing a sufficient number to cover the lung volume. Tumor volume per animal was quantified using 3D Slicer by manual quantification of at least 8 consecutive axial image sequences. MRI was performed to follow tumor volume and weights were monitored bi-weekly. Mice were euthanized and lungs and livers were perfused with 10% formalin, stored in fixative overnight, and embedded in paraffin. For further staining with hematoxylin and eosin (H%E) and antibodies, sections of 5 pm were cut.

Xenograft studies

H69AR xenograft model was established by subcutaneous (s.c.) injection of 2.5 x 10 5 sgCTRL or sgMAVS-H69AR cells in matrigel (Corning, Coming, NY) into the flank of nude mice (Charles River Laboratories, Wilmington, MA). Tumor volume was determined from caliper measurements of tumor length (L) and width (W) according to the formula L x W 2 /2. Both tumor size and body weight were measured three times per week.

Immunoblotting, antibodies and ELISA

Protein was isolated from cell lines and measured by BCA (Pierce Biotechnology). Protein extracts were subjected to polyacrylamide gel electrophoresis using the 4%-l2% NuPAGE gel system (Invitrogen, Carlsbad, CA), transferred to PVDF (Millipore) membranes, and immunoblotted using antibodies that specifically recognize TBK1 (#3013), S172 pTBKl (#5483), pERKl/2 (#4370), ERK1/2 (#9107), S473 pAKT (#4060), AKT (#9272), S396 pIRF3 (#4947), IRF3 (#4302), Y701 pSTATl (#9171), STAT1 (#9172), AXL (#8661), EZH2 (#5246), STING (#13647), MAVS (#3993), E-Cadherin (#3195), Vimentin (#5741), b-Actin (#4970), Tubulin (#2144) (Cell Signaling Technologies, Danvers, MA) and IKKe (#14907) (Sigma- Aldrich, St. Louis, MO).

Secondary antibodies were from LICOR Biosciences (Lincoln, NE): IRDye 800CW Goat anti-Mouse IgG (H + L) (#926-32210), IRDye 800CW Goat anti-Rabbit IgG (H + L) (#926-32211). LICOR blocking buffer (#927-40000) was used to dilute primary and secondary antibodies, with the exception of phosho-specific antibodies, which were diluted in HIKARI Signal Enhancer Solutions 1 and Solution 2 (Nacalai USA, Inc. # NU00101). Imaging of blots and quantitation of bands was performed using the LICOR Odyssey system.

Proteome Profiler™ Human Cytokine Array Kit (#ARY005B) and CXCL10 ELISA (#DIPl00) (R&D Systems, Minneapolis, MN), were performed according to manufacturer’s instructions. For cytokine array, conditioned media (CM) from SCLC cells at basal conditions was collected after 48 and 72 hours. For CXCL10 ELISA, CM from IFN-g pulsed cells was collected after 72 hours.

Microfluidic devices design and fabrication

Microfluidic device design and fabrication was performed as described 27 , with modifications of device dimensions to accommodate larger volumes of media. DAX-l 3D cell culture chip (AIM Biotech, Singapore) was also used for select studies.

Microfluidic culture

H69, H69M and H69AR cell suspensions (2.5 x 10 4 cells) were pelleted and resuspended in type I rat tail collagen (Coming, Coming, NY) at a concentration of 2.5 mg/mL following addition of lOx PBS with phenol red with pH adjusted using NaOH. pH 7.0-7.5 confirmed using PANPEHA Whatman paper (Sigma-Aldrich, St. Louis, MO). The cell-collagen mixture was then injected into the center gel region of the 3D microfluidic culture device. Microfluidic culture devices were designed with a central region containing the cell-collagen mixture, surrounded by 2 media channels located on either side formed by bonding a coverslip to a patterned polydimethylsiloxane (PDMS) substrate. Collagen hydrogels containing cells were incubated 30 minutes at 37°C and then hydrated with media with or without 2.5 x 10 4 cells CFSE labeled Jurkat T cells and THP1 monocytes in the side media channels. Jurkat T cells or THP1 monocytes were labeled with the CFSE Cell Division Tracker Kit (BioLegend, San Diego, CA) following manufacturer’s instructions. Following 48 hours of incubation, images were captured on a Nikon Eclipse 80i fluorescence microscope equipped with Z-stack (Prior) and CoolSNAP CCD camera (Roper Scientific). Image capture and analysis was performed using NIS-Elements AR software package. Whole device images were achieved by stitching in multiple captures. Cell quantitation was performed by measuring total cell area of CFSE dye.

Ex vivo culture of patient-derived organotypic tumor spheroids (PDOTS)

PDOTS from human NSCLC resection specimens were generating according to the recent publication 21 . Briefly, fresh tumor specimens were minced and resuspended in media with collagenase type IV (Life Technologies, Carlsbad, CA). After digestion, samples were strained over 100 pm filter and 40 pm filters to generate Sl (>100 pm), S2 (40-100 pm), and S3 (<40 pm) spheroid fractions. S2 spheroid fraction was pelleted and resuspended in type I rat tail collagen (Coming, Corning, NY) at a concentration of 2.5 mg/mL. The spheroid-collagen mixture was then injected into the center gel region of the 3D microfluidic culture device. Collagen hydrogels containing PDOTS were hydrated with media and treated with anti-PD-l (Nivolumab, 100 pg/mL), IFNy (200 ng/mL), Poly (I:C) (0.5 pg/mL) or combination (Nivolumab + IFNy Nivolumab + Poly (I:C)). The number of Live/Dead cells in each treatment condition was determined by Trypan Blue Exclusion method and plotted as percentage of viability.

Flow cytometry analysis and cell sorting

Cells were stained with anti-PD-Ll (Biolegend, Cat# 329717), anti-CD44 (Biolegend, Cat# 103011) or isotype IgG control antibodies (Biolegend, Cat# 400326 and Cat#4006l l), in PBS containing 2% FBS for analysis and cell sorting. Briefly, cells were washed and further incubated with the indicated antibodies at 2 pg/mL. After 3 washes with PBS, cells were resuspended in PBS containing 2% FBS and analyzed on BD FACSCanto II or sorted to >95% purity using a BD FACS Aria II. Levels were compared with isotype control antibodies. PD-L1 and CD44 mean fluorescence intensity (MFI) was normalized to isotype control. The data analyses were performed with FlowJo software (TreeS tar). Cytokine profiling

Multiplex assays were performed utilizing the bead-based immunoassay approach Bio- Plex Pro™ Human Cytokine 40-plex Assay (Cat# 171AK99MR2) on a Bio-plex 200 system (Cat# 171000201) (Bio-Rad Laboratories, Hercules, CA) and the Human Cytokine/Chemokine Maganetic Bead Panel (Cat# HCYTMAG-60K-PX30) on a Luminex MAGPIX system (Merck Millipore, Billerica, MA). Conditioned media concentration levels [pg/ml] of each protein were derived from 5- parameter curve fitting models. Fold changes relative to the corresponding control were calculated and plotted as log2FC. Lower and upper limits of quantitation (LLOQ/ULOQ) were imputed from standard curves for cytokines above or below detection.

Quantitative RT-PCR

Total cellular RNA was extracted using the miRNeasy Mini Kit (Qiagen, Hilden, Germany) according to manufacturer’s instructions. After extraction, 1 pg total RNA was used to generate cDNA with the Superscript III First-Strand Synthesis SuperMix for qRT-PCR kit (Thermo Fisher Scientific, Waltham, MA). Quantitative reverse transcription PCR (qRT-PCR) of the indicated genes was performed using SYBR green PCR Master Mix (Applied Biosystems, Foster City, CA) and the Applied Biosystems 7300 Fast real-time PCR system and software. The relative expression was normalized with the expression of the housekeeping gene 36B4. The sequences of primers used have been listed in Table 3.

Table 3: Gene primer sequences

Table 4: ERV Primer Sequences dsRNA enrichment

For dsRNA enrichment, RNA was first treated or not for 30 minutes with 50pg/ml RNase A (Qiagen, Hilden, Germany) in high-salt concentration (NaCl, 0.35 M) to prevent dsRNA degradation. After treatment, RNase A was removed by ethanol precipitation and the product was resuspended in sterile water. Next, RNase A-treated RNA was immunoprecipitated (IP) using the J2 dsRNA-specific antibody (English and Scientific Consulting Kft, Szirak, Hungary). In brief, the product of 9 pg of RNase A-treated RNA was incubated in binding buffer (150 mM NaCl, 50 mM TRIS pH8.0, 1 mM EDTA, 1% NP-40) with 5 pg of J2 antibody rotating overnight at 4°C. J2-bound dsRNA was incubated in biding buffer with 25 pL of Dynabeads Protein G (Thermo Fisher Scientific, Waltham, MA) for 4 hours at 4°C, followed by 5 washes in cold binding buffer. RNA was then extracted with TRIzol Reagent and expression levels of indicated genes were analyzed by qRT-PCR.

Enrichment of dsRNA over ssRNA was then calculated by normalizing the delta Ct of ERVs (dsRNA) against beta-actin (ssRNA).

First strand cDNA synthesis and strand specific PCR for detection of sense and antisense ERV transcripts using TASA-TD methodology Components from the Superscript III First-Strand Synthesis System for RT-PCR (Thermo Fisher Scientific, Waltham, MA) were adapted to perform reverse transcription with RNA from H69AR previously pulsed with IFNy 10 minutes. For first strand cDNA synthesis 400 ng RNA was used for B-actin, MLT1C49, MLT1A and MLT1J. ImM of a gene specific primer ligated to a TAG-sequence not specific for the human genome (GSP sense/antisense (RT) TAG) was implemented in the reaction. RNA and primers were preheated at 65°C for 5min. For the total reaction: the GSP-TAG, 0.5mM dNTP, 5mM MgCl2, lOmM DTT, 40U RNaseOUT, 100U SuperS crip till® RT and 240ng Actinomycin D (Sigma-Aldrich, St. Louis, MO) were added with the RNA for a 20pl reaction. Synthesis was performed at 50°C for 50 minutes and terminated at 85°C for 5 min. RT with extremely low intrinsic RNase H activity (for cleavage of RNA from RNA/DNA duplexes) and Actinomycin D was added to prevent second strand cDNA RT resulting in antisense artifacts 28 . After cDNA synthesis 2U recombinant RNase H was added to each reaction and incubated 20 minutes at 37°C. Finally, the first strand cDNA mix was ethanol precipitated and resuspended in 10 mΐ sterile water. Afterwards gene and strand specific qRT-PCR was performed using SYBR green PCR Master Mix. To amplify sense cDNA and antisense cDNA a TAG-primer and GSP sense-primer and a TAG-primer and GSP antisense- primer were used, respectively. Sense and antisense specific qRT-PCR was performed using both sense and antisense cDNA of beta-actin as an internal negative control that was previously demonstrated to have no antisense transcript 29 . The sequences of primers used have been listed in Table 5.

Table 5: Primers used for TASA-TD method

CRISPR-Cas9 gene editing and lentiviral infection

Oligonucleotides coding for guide RNAs that target STING and MAVS genes were chosen from the Avana library and the Brunello library 30 . A non-targeting sgRNA from the Gecko library v2 was used as a dummy sgRNA for control 31 . Lenti CRISPRv2 vectors were cloned as previously described 31,32 . sgRNA target sequences are described on Table 6.

HEK-293T cells were transduced with lentiCRISPRv2 using X-treme Gene 9 (Roche, Basel, Switzerland) according to the manufacturer's instructions. On day 2, target cells were seeded, and allowed to adhere overnight. On day 3 the supernatant of transduced HEK293T cells was collected and added to the target cells through a 0.45 pm filter. Supernatant from transduced HEK293T cells was again collected and added to target cells on day 4. On day 5, puromycin or blasticidin was added to select infected cells (for four days).

Table 6: CRISPR-Cas9 sgRNAs target sequences

Poly(I:C) treatment

For Poly(LC) dsRNA treatment experiments, H69ARsgCTRL and H69ARsgMAVS cells were plated in RPMI media, transfected with 0.5 pg/mL Poly(LC) HMW (InvivoGen, Sand Diego, CA) using XtremeGene HP transfection reagent (Sigma-Aldrich, St. Louis, MO) and cultured for 72 hours. On day 3 after transfection, conditioned media was recovered and CXCL10 protein expression was quantified using Human CXCL10/IP-10 Quantikine ELISA kit (R&D Systems, Minneapolis, MN). RNA was extracted and expression levels of relevant genes were analyzed by qRT-PCR.

Immunohistochemical staining

After deparaffinizing tissue blocks, antigen retrieval was achieved by wet autoclave (121 degrees Celsius, 15 min) in Antigen Retrieval Solution, pH 9 (Dako, S2367) for p-TBKl. In order to block endogenous peroxide enzyme, tissue sections were incubated for 30 minutes using Peroxidase-Blocking Solution (Dako, S2023). Then, to block non-specific background staining, tissue sections were incubated for 20 minutes with Protein Block (Dako, X0909) (human tissue) or Mouse on Mouse blocking reagent (Vector Laboratories, MKB-2213) (mouse tissue). Primary antibody specific for pTBKl (Cell Signaling Technology, 5483; 1:50 dilution) was applied, and slides were incubated for 16 hours at 4°C. Visualization was achieved using EnVision™+/HRP, Rabbit (Dako, K4003) for pTBKl, followed by diaminobenzidine (Dako, K3468), and hematoxylin counterstain. Expression levels of pTBKl were evaluated by two pathologists who were blinded to other data.

Multiplexed Immunofluorescence

Multiplex Immunofluore scent staining was performed overnight for approximately 12 hours on BOND RX fully automated Stainers (Leica Biosystems) as previously described 33 . Briefly, tissue sections of 5-pm thick FFPE were baked for 3 hours at 60°C before loading into the BOND RX. Slides were deparaffinized (BOND DeWax Solution, Leica Biosystems) and rehydrated with series of graded ethanol to deionized water. Antigen retrieval was performed in BOND Epitope Retrieval Solution 1 (ER1, Leica Biosystems) at pH 6 for 10 minutes at 98°C. Deparaffinization, rehydration and antigen retrieval were all preprogrammed and executed by the BOND RX. Next, slides were serially stained with primary antibodies, such as anti-CD8 (clone C8/144B; DAKO, dilution 1:5000). Incubation time per primary antibody was 40 minutes. Subsequently, anti-rabbit Polymeric Horseradish Peroxidase (Poly-HRP, BOND Polymer Refine Detection Kit, Leica Biosystems) was applied as a secondary label with an incubation time of 10 minutes. Signal for antibody complexes was labeled and visualized by their corresponding Opal Fluorophore Reagents (PerkinElmer) by incubating the slides for 5 minutes. The same process was repeated for the following antibodies/fluorescent dyes. Slides were air dried, mounted with Prolong Diamond Anti-fade mounting medium (#P36965, Life Technologies) and stored in a light-proof box at 4C prior to imaging. The target antigens, antibody clones, and dilutions for markers included in this report and details of controls are listed in Table 7.

Table 7. Antibodies used for multiplexed immunofluorescence assay.

Table summarizing target antigens, antibody clones, dilutions and detail of controls used for multiplexed immunofluorescence staining of NSCLC human specimens used to generate PDOTS.

Image acquisition and analysis

Image acquisition was performed using the Mantra multispectral imaging platform (Vectra 3, PerkinElmer, Hopkinton, MA) as previously described 33 . Areas with non-tumor or residual normal tissue (i.e. residual lymph node) were excluded from the analysis. Representative regions of interest were chosen by the pathologist, and 5-7 fields of view (FOVs) were acquired at 20x resolution as multispectral images. Image Analysis was performed using the Inform 2.3 Image Analysis Software (PerkinElmer, Hopkinton, MA).

OncoPanel assay Somatic mutations, copy number variations and structural variants in parental H69 cells and H69M-PD-Ll low /H69M-PD-Ll hlgh subclones were evaluated by performing the OncoPanel assay from the Center for Advanced Molecular Diagnostics from Brighman and Women’s Hospital.

This OncoPanel assay surveys exonic DNA sequences of 300 cancer genes and 113 introns across 35 genes for rearrangement detection. DNA was isolated from cell lines and analyzed by massively parallel sequencing using a solution-phase Agilent SureSelect hybrid capture kit and an Illumina HiSeq 2500 sequencer.

The 300 genes are: ABL1, AKT1, AKT2, AKT3, ALK, ALOX12B, APC, AR, ARAF, ARID 1 A, ARID1B, ARID2, ASXL1, ATM, ATRX, AURKA, AURKB, AXL, B2M, BAP1, BCL2, BCL2L1, BCL2L12, BCL6, BCOR, BCORL1, BLM, BMPR1A, BRAF, BRCA1, BRCA2, BRD4, BRIP1, BUB 1B, CADM2, CARD11, CBL, CBLB, CCND1, CCND2, CCND3, CCNE1, CD274, CD58, CD79B, CDC73, CDH1, CDK1, CDK2, CDK4, CDK5, CDK6, CDK9, CDKN1A, CDKN1B, CDKN1C, CDKN2A, CDKN2B, CDKN2C, CEBPA, CHEK2, CIITA, CREBBP, CRKL, CRLF2, CRTC1, CRTC2, CSF1R, CSF3R, CTNNB 1, CUX1, CYLD, DDB2, DDR2, DEPDC5, DICER1, DIS3, DMD, DNMT3A, EED, EGFR, EP300, EPHA3, EPHA5, EPHA7, ERBB2, ERBB3, ERBB4, ERCC2, ERCC3, ERCC4, ERCC5, ESR1, ETV1, ETV4, ETV5, ETV6, EWSR1, EXT1, EXT2, EZH2, FAM46C, FANCA, FANCC, FANCD2, FANCE, FANCF, FANCG, FAS, FBXW7, FGFR1, FGFR2, FGFR3, FGFR4, FH, FKBP9, FLCN, FLT1, FLT3, FLT4, FUS, GATA3, GATA4, GATA6, GLI1, GLI2, GLI3, GNA11, GNAQ, GNAS, GNB2L1, GPC3, GSTM5, H3F3A, HNF1A, HRAS, ID3, IDH1, IDH2, IGF1R, IKZF1, IKZF3, INSIG1, JAK2, JAK3, KCNIP1, KDM5C, KDM6A, DM6B, KDR, KEAP1, KIT, KRAS,LINC00894, LMOl, LM02, LM03, MAP2K1, MAP2K4, MAP3K1, MAPK1, MCL1, MDM2, MDM4, MECOM, MEF2B, MEN1, MET, MITF, MLH1, MLL(KMT2A), MLL2(KTM2D), MPL, MSH2, SH6, MTOR, MUTYH, MYB, MYBL1, MYC, MYCLl(MYCL), MYCN, MYD88, NBN, NEGR1, NF1, NF2, NFE2L2, NFKBIA, NFKBIZ, NKX2-1, NOTCH1, NOTCH2, NPM1, NPRL2, NPRL3, NRAS, NTRK1, NTRK2, NTRK3, PALB2, PARK2, PAX5, PBRM1, PDCD1LG2, PDGFRA, PDGFRB, PHF6, PHOX2B, PIK3C2B, PIK3CA, PIK3R1, PIM1, PMS 1, PMS2, PNRC1, PRAME, PRDM1, PRF1, PRKAR1A, PRKCI, PRKCZ, PRKDC, PRPF40B, PRPF8, PSMD13, PTCH1, PTEN, PTK2, PTPN11, PTPRD, QKI, RAD21, RAF1, RARA, RB 1, RBL2,RECQL4, REL, RET, RFWD2, RHEB, RHPN2, ROS 1, RPL26, RUNX1, SBDS, SDHA, SDHAF2, SDHB, SDHC, SDHD, SETBP1, SETD2, SF1, SF3B 1, SH2B3, SLITRK6, SMAD2, SMAD4, SMARCA4, SMARCB 1, SMC1A, SMC3, SMO, SOCS 1, SOX2, SOX9, SQSTM1, SRC, SRSF2, STAG1, STAG2, STAT3, STAT6, STK11, SUFU, SUZ12, SYK, TCF3, TCF7L1, TCF7L2, TERC, TERT, TET2, TLR4, TNFAIP3, TP53, TSC1, TSC2, U2AF1, VHL, WRN, WTl,XPA, XPC, XPOl, ZNF217, ZNF708, ZRSR2.

Intronic regions are tiled on specific introns of ABL1, AKT3, ALK, BCL2, BCL6, BRAF, CUT A, EGFR, ERG, ETV1, EWSR1, FGFR1, FGFR2, FGFR3, FUS, IGH, IGL, JAK2, MLL, MYC, NPM1, NTRK1, PAX5, PDGFRA, PDGFRB, PPARG, RAF1, RARA, RET, ROS 1, SS 18, TRA, TRB, TRG, TMPRSS2.

ATAC-sequencing and analysis

ATAC sequencing was performed on H69 and H69AR cells according to 34 .

Briefly, 40-50,000 cells were sorted per biological replicate, which were then washed once in cold PBS and lysed in 50pL cold lysis buffer (10 mM Tris-HCl, pH 7.4, 10 mM NaCl, 3 mM MgCl2, 0.1% IGEPAL CA-630). Lysed nuclei were incubated in Tn5 transposition reaction mix and purified using MinElute Reaction Cleanup kit (Qiagen). ATAC-seq fragments from one set of replicates for H69 and H69AR cells were size selected for fragments between 115 and 600 bp using Pippin Prep 2% Agarose Gel Cassettes and the Pippin Prep DNA Size Selection System (Sage Science). Post size- selection, ATAC libraries were amplified and Nextera sequencing primers ligated using Polymerase Chain Reaction (PCR). Finally, PCR primers were removed using Agencourt AMPure XP bead cleanup (Beckman Coulter/ Agencourt) and library quality was verified using a Tapestation machine. High quality ‘multiplexed’ DNA libraries were sequenced on the Illumina HiSeq2000.

The ends of the paired-end fragments are used as cut sites and enriched peaks were called with MACS2 with following parameters (—nomodel — extsize 200—shift -100 -g hs -B— nolambda). For IGV visualization, shifted bedGraph were converted to wig files at lObp resolution and normalized to read counts by wigmath tool of JavaGenomic toolkit.

Genomic analysis and SPARCS gene set derivation

The H69 vs H69M expression microarray data analyzed in this study were obtained from the publicly available dataset GSE45120. ChlP-seq data for STAT1 and IRF1 data on CD 14+ monocytes were retrieved from GEO database (GSE43036). Replicates data for each ChIP were aggregated into a single wiggle file and visualized in IGV genome browser at the locus of TRIM22 gene 35 .

Genomic coordinates of all 3’UTR from NCBI RefSeq transcripts were intersected with all repeat elements that are 50bp or longer and have its family name containing a string‘ERV’ from UCSC Repeat Masker. Those 5880 3’UTRs that overlap with any ERVs were collapsed to the gene level. Those 1,080 unique genes were further used to find overlaps with differentially expressed gene from an analysis comparing H69M vs H69. Of 452 significantly upregulated genes (adjP<lE-4 and logFC>2) in H69M, 22 genes were found overlapped. As only 86 genes were identified at the same threshold, the same number of genes (452) most significantly downregulated genes at logFC l were used to find overlaps with the genes containing ERV in its 3’ UTR (25 genes).

Gene set enrichment analysis

SPARCS signature was created by overlapping 3’UTR repeat elements from Ref Seq with genes upregulated in H69M when compared to parental H69 5 . The SPARCS gene set scores across CCLE (broadinstitute.org/CCLE) or TCGA (Pancanl2) 17 were derived by using ssGSEA algorithm 36 . These scores were used to identify top associated features from the matching datasets containing profiles of mRNA, pathways (MsigDB), and mutations/copy number variation data (broadinstitute.org/CCLE). To quantify the degree of association, an information-theoretic measure Information Coefficient (IC) 18 was used and an empirical permutation test for statistical significance calculations.

Statistical analyses

All graphs depict mean + s.d. unless otherwise indicated. Tests for differences between two groups were performed using two-tailed unpaired Student’s /-test or Mann- Whitney’s two- tailed test, as specified in the figure legends. Two-way ANOVA with a Sidak post hoc test was performed to compare tumor growth between groups in xenograft studies. P values were considered significant if less than 0.05. Asterisks used to indicate significance correspond with: *p<0.05, **p<0.0l, ***p<0.00l. GraphPad Prism7 was used for statistical analysis of experiments, data processing and presentation. References

1 Hanahan, D. & Weinberg, R. A. Hallmarks of cancer: the next generation. Cell 144, 646- 674, doi: 10. l0l6/j.cell.20l 1.02.013 (2011).

2 Marusyk, A. el al. Non-cell- autonomous driving of tumour growth supports sub-clonal heterogeneity. Nature 514, 54-58, doi:l0.l038/naturel3556 (2014).

3 Junttila, M. R. & de Sauvage, F. J. Influence of tumour micro-environment heterogeneity on therapeutic response. Nature 501, 346-354, doi: 10.1038/nature 12626 (2013).

4 Tabassum, D. P. & Polyak, K. Tumorigenesis: it takes a village. Nat Rev Cancer 15,

473-483, doi: 10. l038/nrc3971 (2015).

5 Canadas, I. et al. Targeting epithelial-to-mesenchymal transition with Met inhibitors reverts chemoresistance in small cell lung cancer. Clin Cancer Res 20, 938-950, doi:l0.H58/l078-0432.CCR-l3-l330 (2014).

6 Calbo, J. et al. A functional role for tumor cell heterogeneity in a mouse model of small cell lung cancer. Cancer Cell 19, 244-256, doi: 10. l0l6/j.ccr.20l0.12.021 (2011).

7 Konieczkowski, D. J. et al. A melanoma cell state distinction influences sensitivity to MAPK pathway inhibitors. Cancer Discov 4, 816-827, doi:l0.H58/2l59-8290.CD-l3- 0424 (2014).

8 Tirosh, I. et al. Dissecting the multicellular ecosystem of metastatic melanoma by single cell RNA-seq. Science 352, 189-196, doi:l0.H26/science.aad050l (2016).

9 Crescenzo, R. et al. Convergent mutations and kinase fusions lead to oncogenic STAT3 activation in anaplastic large cell lymphoma. Cancer Cell 27, 516-532, doi : 10.1016/j . ccell .2015.03.006 (2015).

10 Wu, X. et al. AXL kinase as a novel target for cancer therapy. Oncotarget 5, 9546-9563, doi:l0.l8632/oncotarget.2542 (2014).

11 Chiappinelli, K. B. et al. Inhibiting DNA Methylation Causes an Interferon Response in

Cancer via dsRNA Including Endogenous Retroviruses. Cell 162, 974-986, doi: 10. l0l6/j.cell.20l5.07.011 (2015).

12 Roulois, D. et al. DNA-Demethylating Agents Target Colorectal Cancer Cells by

Inducing Viral Mimicry by Endogenous Transcripts. Cell 162, 961-973, doi: 10. l0l6/j.cell.20l5.07.056 (2015).

13 Chuong, E. B., Elde, N. C. & Feschotte, C. Regulatory evolution of innate immunity through co-option of endogenous retroviruses. Science 351, 1083-1087, doi: 10.1 l26/science.aad5497 (2016).

14 Gao, D. et al. Cyclic GMP-AMP synthase is an innate immune sensor of HIV and other retroviruses. Science 341, 903-906, doi: 10. H26/science.1240933 (2013). Poirier, J. T. el al. DNA methylation in small cell lung cancer defines distinct disease subtypes and correlates with high expression of EZH2. Oncogene 34, 5869-5878, doi:l0.l038/onc.20l5.38 (2015).

Henke, C. et al. Selective expression of sense and antisense transcripts of the sushi-ichi- related retrotransposon— derived family during mouse placentogenesis. Retrovirology 12, 9, doi: 10. H86/s 12977-015-0138-8 (2015).

Hoadley, K. A. et al. Multiplatform analysis of 12 cancer types reveals molecular classification within and across tissues of origin. Cell 158, 929-944, doi:l0.l0l6/j.cell.20l4.06.049 (2014).

Kim, J. W. et al. Characterizing genomic alterations in cancer by complementary functional associations. Nat Biotechnol 34, 539-546, doi:l0.l038/nbt.3527 (2016).

Rhodes, D. A., Reith, W. & Trowsdale, J. Regulation of Immunity by Butyrophilins. Annu Rev Immunol 34, 151-172, doi:l0.H46/annurev-immunol-04l0l5-055435 (2016).

Bindea, G. et al. Spatiotemporal dynamics of intratumoral immune cells reveal the immune landscape in human cancer. Immunity 39, 782-795, doi: 10. l0l6/j.immuni.2013.10.003 (2013).

Jenkins, R. W. et al. Ex Vivo Profiling of PD-l Blockade Using Organotypic Tumor Spheroids. Cancer Discov, doi: 10.1158/2159-8290.CD- 17-0833 (2017).

Pan, D. et al. A major chromatin regulator determines resistance of tumor cells to T cell- mediated killing. Science , doi:lO.H26/science.aaol7lO (2018).

Miao, D. et al. Genomic correlates of response to immune checkpoint therapies in clear cell renal cell carcinoma. Science , doi:lO.H26/science.aan595l (2018).

Manguso, R. T. et al. In vivo CRISPR screening identifies Ptpn2 as a cancer immunotherapy target. Nature 547, 413-418, doi:l0.l038/nature23270 (2017).

Peng, D. et al. Epigenetic silencing of THl-type chemokines shapes tumour immunity and immunotherapy. Nature 527, 249-253, doi: 10.1038/nature 15520 (2015).

Christensen, C. L. et al. Targeting transcriptional addictions in small cell lung cancer with a covalent CDK7 inhibitor. Cancer Cell 26, 909-922, doi: 10. l0l6/j.ccell.20l4.10.019 (2014).

Aref, A. R. et al. Screening therapeutic EMT blocking agents in a three-dimensional microenvironment. Integr Biol (Camb) 5, 381-389, doi:l0.l039/c2ib20209c (2013).

Perocchi, F., Xu, Z., Clauder-Munster, S. & Steinmetz, L. M. Antisense artifacts in transcriptome microarray experiments are resolved by actinomycin D. Nucleic Acids Res 35, el28, doi:l0.l093/nar/gkm683 (2007).

Chen, J. et al. Over 20% of human transcripts might form sense-antisense pairs. Nucleic Acids Res 32, 4812-4820, doi:l0.l093/nar/gkh8l8 (2004). 30 Doench, J. G. et al. Optimized sgRNA design to maximize activity and minimize off- target effects of CRISPR-Cas9. Nat Biotechnol 34, 184-191, doi:l0.l038/nbt.3437 (2016).

31 Sanjana, N. E., Shalem, O. & Zhang, F. Improved vectors and genome- wide libraries for CRISPR screening. Nat Methods 11, 783-784, doi:l0.l038/nmeth.3047 (2014).

32 Shalem, O. et al. Genome-scale CRISPR-Cas9 knockout screening in human cells.

Science 343, 84-87, doi: 10. H26/science.1247005 (2014).

33 Carey, C. D. et al. Topological analysis reveals a PD-L1 -associated microenvironmental niche for Reed-Sternberg cells in Hodgkin lymphoma. Blood 130, 2420-2430, doi:l0.H82/blood-20l7-03-7707l9 (2017).

34 Sen, D. R. et al. The epigenetic landscape of T cell exhaustion. Science 354, 1165-1169, doi : 10.1126/science . aae0491 (2016).

35 Qiao, Y. et al. Synergistic activation of inflammatory cytokine genes by interferon- gamma-induced chromatin remodeling and toll-like receptor signaling. Immunity 39, 454-469, doi: 10. l0l6/j.immuni.20l3.08.009 (2013).

36 Barbie, D. A. et al. Systematic RNA interference reveals that oncogenic KRAS-driven cancers require TBK1. Nature 462, 108-112, doi:l0.l038/nature08460 (2009).

Example 2: STING Levels

STING levels vary widely in KRAS mutant lung cancer and correlate with LKB1 and DNMT1 status

KRAS mutant non-small cell lung cancer (NSCLC) is a heterogeneous disease, and co mutation of the tumor suppressor genes p53 or LKB1 defines different subtypes. KRAS;p53 mutant (KP) or KRAS;LKB1 mutant (KL) NSCLC cells exhibit divergent gene expression profiles and sensitivity to immune therapies. For example, KL NSCLC showed significantly lower expression of immune checkpoint molecules such as PD-L1 compared with KP NSCLC, implying less efficacy to PD-L1 targeted therapy in KL NSCLC patients (2, 3)

Using the Cancer Cell Line Encyclopedia (CCLE) database (4), differentially expressed genes between KP and KL cell lines were first extracted. Of those genes, 48 genes were highly expressed in KP cell lines, and also annotated as immune-related genes based on Gene Ontology (GO) term. TMEM173/STING (hereafter STING) is a critical regulator of innate immune response to cytosolic double strand DNA, and one of the top-ranked genes in this list. It was next demonstrated that STING expression was higher in KP cell lines in both mRNA and protein levels (Figures 15A and 15B). Since KRAS/LKBl/p53 mutant cells also showed lower expression compared with KP cells (HCC44, H2122, H1355, H23), these data suggested that the dominant feature associated with STING expression was LKB1 as opposed to p53 mutation status (loss of LKB1 = low STING levels).

Given these data, it was next examined whether LKB1 directly regulates STING expression in KRAS mutant lung cancer. Interestingly, LKB1 reconstitution clearly increased STING expression in specific KL cell lines such as H1944, H2122 and H1355 (Figure 16A). On the other hand, other KL cell lines such as A549, A427 and HCC44 were insensitive to LKB 1 reconstitution (Figure 16A). Since LKB1 loss leads to hyperactive DNA methyltransferase (DNMT) activity (5), which could silence STING epigenetic ally, it was wondered whether baseline DNMT1 expression might modulate this relationship in KL cells. Indeed, KL cell lines that were insensitive to LKB1 reconstitution expressed higher levels of DNMT1 than the sensitive lines (Figure 16B). Interestingly, LKB 1 reconstitution in the presence of a DNMT inhibitor in A549 cells (KL DNMT high cell line) was able to restore STING expression (Figure 16C). Together, these data reveal that (1) LKB 1 directly up-regulates STING expression in KRAS mutant lung cancer cells and (2) STING gene can be silenced by DNA methylation, and that DNA hypomethylation of the STING gene locus is required for LKB1 to regulate STING expression.

It was next examined whether increased STING expression following LKB1 reconstitution in KL cells altered their innate immune responsiveness to dsDNA. Transfection of a synthetic analogue of B-DNA, poly(dA:dT), strongly activated STING downstream pathway including TBK1 and IRF3 in LKB1 reconstituted KL cells compared with control cells (Figure 17A). Poly(dA:dT) stimulation also promoted secretion of CXCL10, a key IRF3 target, specifically in LKB1 reconstituted cells (Figure 17B). These results confirm that increased STING expression following LKB1 reconstitution contribute to enhanced innate immune response to cytosolic double-strand DNA.

Regulation of STING by EZH2 in small cell lung cancer

More broadly, epigenetic silencing, including regulation of specific histone modifications, is increasingly recognized in addition to DNA methylation as an important determinant in cancer immunology and immunotherapy response. For example, EZH2, which promotes histone H3K27 trimethylation and silencing of gene transcription, has been shown to play a key role in regulating Thl chemokine expression and CD8(+) T cell infiltration in certain tumors (6), although the mechanism has been incompletely characterized. Interestingly, recent studies have implicated Endogenous Retrovirus (ERV) reactivation following DNA methyltransferase (DNMT) as a specific mechanism that may prime tumor immune responses through recognition of dsRNA and potentially dsDNA, suggesting a potentially synergistic approach to enhance immunogenicity in combination with PD-l immune checkpoint blockade (7, 8).

Along these lines, by exploiting a human Small Cell Lung Cancer Model (SCLC) that exhibits tumor cell heterogeneity (H69 cells), an epigenetically regulated subclass of ERVs that promotes pathologic innate immune signaling, termed“Stimulated 3 Prime Antisense Retroviral Coding Sequences” (SPARCS) was recently identified (Canadas et ah, Nature Medicine, in revision). These ERVs are poised to undergo positive feedback signal amplification due to their antisense localization in 3’ UTRs of IFN stimulated genes, resulting dsRNA production, viral sensing, and positive feedback induction of their expression by IFN. Whereas neuroendocrine SCLC cells and other epithelial cancer cell lines silence these genes, mesenchymal subclones with high AXL/MET expression and low EZH2 levels are particularly susceptible to triggering SPARCS expression when exposed to IFNy. Furthermore, it was directly demonstrated that these ERVs are silenced by EZH2, since treatment with an EZH2 inhibitor (GSK126) induced their reactivation in parental neuroendocrine H69 cells.

Given the above observations differences in STING levels between the parental neuroendocrine cell line H69 and the derivative drug-resistant subclones H69M and H69AR were also examined. Interestingly it was found that, in contrast to H69 cells, the resistant subclones expressed high levels of STING, as well as the H69M-PD-Ll hlgh subpopulation (Figure 18). ETsing ATAC seq and H3K27Ac ChIP seq the development of peaks in the 5’ promoter region of TMEM173 as well as 3’ enhancer in these tumor cell subclones relative to the parental H69 cell line was further confirmed.

Variability of STING expression across human cancer types

To determine the broader relevance of STING expression in cancer, as well as specific features linked to the SPARCS high state, STING expression across SPARCS hlgh and SPARCS low cell lines was explored using the Cancer Cell Line Encyclopedia (CCLE) database. Interestingly, a robust and significant association of SPARCS hlgh cell lines with high levels of STING was observed when compared with SPARCS low cell lines (Figure 20A). 16 cell lines with high, intermediate, or low SPARCS signature expression were therefore selected and STING protein levels were determined. Of note, SPARCS hlgh cells tightly correlated with particularly high protein expression of STING compared with a total lack of expression of this protein in SPARCS low cells (Figure 20B). The only exception among the SPARC hlgh cell lines that failed to express STING was HCC44, one of the KL cell lines lacking LKB1 and with high DNMT expression (Figure 16).

Finally, it was observed that within this larger panel of cell lines, triple negative breast cancer (TNBC) cells (MDA-MB-231, HCC1143, MDA-MB-468) were STING positive, whereas estrogen receptor positive (ER+) cells (T47D, MDA-MB-453) were STING negative. Therefore a larger panel of breast cancer cell lines was characterized to determine if this trend held up. Indeed, whereas 5/6 TNBC cell expressed detectable levels of STING (1569, 1143, 70, 468, and 231), all 3 ER+ cell lines were STING negative (CAMA-l, T47D, and MCF7). These data suggest that TNBC in particular is a tumor type in which focused examination of STING levels in tumors and deployment of STING agonists +/- PD-l blockade may have significant therapeutic impact. As above, it is also likely that epigenetic inhibitors (e.g. DNMT or EZH2) may convert STING 0W TNBC tumors or ER+ cell lines into STING hlgh and prime them to respond to STING directed immunotherapy approaches.

References:

1. Barber GN. STING: infection, inflammation and cancer. Nat Rev Immunol.

2015;15(12):760-70.

2. Skoulidis F, Byers LA, Diao L, Papadimitrakopoulou VA, Tong P, Izzo J, Behrens C, Kadara H, Parra ER, Canales JR, et al. Co-occurring Genomic Alterations Define Major Subsets of KRAS-Mutant Lung Adenocarcinoma with Distinct Biology, Immune Profiles, and Therapeutic Vulnerabilities. Cancer Discov. 20l5;5(8):860-77.

3. Koyama S, Akbay EA, Li YY, Aref AR, Skoulidis F, Herter-Sprie GS, Buczkowski KA, Liu Y, Awad MM, Denning WL, et al. STK11/LKB1 deficiency promotes neutrophil recruitment and proinflammatory cytokine production to suppress T cell activity in the lung tumor microenvironment. Cancer Res. 2016.

4. Barretina J, Caponigro G, Stransky N, Venkatesan K, Margolin AA, Kim S, Wilson CJ, Lehar J, Kryukov GV, Sonkin D, et al. The Cancer Cell Line Encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 20l2;483(739l):603-7.

5. Kottakis F, Nicolay BN, Roumane A, Karnik R, Gu H, Nagle JM, Boukhali M, Hayward MC, Li YY, Chen T, et al. LKB1 loss links serine metabolism to DNA methylation and tumorigenesis. Nature. 20l6;539(7629):390-5. 6. Peng D, Kryczek I, Nagarsheth N, Zhao L, Wei S, Wang W, Sun Y, Zhao E, Vatan L, Szeliga W, et al. Epigenetic silencing of THl-type chemokines shapes tumour immunity and immunotherapy. Nature. 20l5;527(7577):249-53.

7. Chiappinelli KB, Strissel PL, Desrichard A, Li H, Henke C, Akman B, Hein A, Rote NS, Cope LM, Snyder A, et al. Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses. Cell. 20l5;l62(5):974-86.

8. Roulois D, Loo Yau H, Singhania R, Wang Y, Danesh A, Shen SY, Han H, Liang G, Jones PA, Pugh TJ, et al. DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts. Cell. 20l5;l62(5):96l-73.

Example 3: STING Levels in Breast Cancer Subtypes

STING protein expression was measured in different breast cancer cell subtypes by western blot analysis on protein extracts from the breast cancer cell lines. Western results are shown in Figure 22A and 22C. STING protein levels are shown relative to b-actin control protein levels. Genotypes of the cell lines (ER +/-, PR+/-, and HER2+/-) are shown in Figure 22B. The data presented herein demonstrates that STING levels are generally increased in TNBC (Triple Negative Breast Cancer) and luminal B cancer cell subtypes.

TNBC cell lines were also assayed for their responsiveness to the STING agonist AduroSlOO. Cells were treated with AduroSlOO or a control and CXCL10 levels secreted into the supernatant were measured. As is shown in Figure 22D, all TNBC cell lines showed elevated levels of the CXCL10 chemokine when treated with AduroSlOO relative to a control, indicating that TNBC cells are responsive to treatment with a STING agonist, regardless of STING levels.

EQUIVALENTS

While several inventive embodiments have been described and illustrated herein, those of ordinary skill in the art will readily envision a variety of other means and/or structures for performing the function and/or obtaining the results and/or one or more of the advantages described herein, and each of such variations and/or modifications is deemed to be within the scope of the inventive embodiments described herein. More generally, those skilled in the art will readily appreciate that all parameters, dimensions, materials, and configurations described herein are meant to be exemplary and that the actual parameters, dimensions, materials, and/or configurations will depend upon the specific application or applications for which the inventive teachings is/are used. Those skilled in the art will recognize, or be able to ascertain using no more than routine experimentation, many equivalents to the specific inventive embodiments described herein. It is, therefore, to be understood that the foregoing embodiments are presented by way of example only and that, within the scope of the appended claims and equivalents thereto, inventive embodiments may be practiced otherwise than as specifically described and claimed. Inventive embodiments of the present disclosure are directed to each individual feature, system, article, material, kit, and/or method described herein. In addition, any combination of two or more such features, systems, articles, materials, kits, and/or methods, if such features, systems, articles, materials, kits, and/or methods are not mutually inconsistent, is included within the inventive scope of the present disclosure.

All definitions, as defined and used herein, should be understood to control over dictionary definitions, definitions in documents incorporated by reference, and/or ordinary meanings of the defined terms.

All references, patents and patent applications disclosed herein are incorporated by reference with respect to the subject matter for which each is cited, which in some cases may encompass the entirety of the document.

The indefinite articles“a” and“an,” as used herein in the specification and in the claims, unless clearly indicated to the contrary, should be understood to mean“at least one.”

The phrase“and/or,” as used herein in the specification and in the claims, should be understood to mean“either or both” of the elements so conjoined, i.e., elements that are conjunctively present in some cases and disjunctively present in other cases. Multiple elements listed with“and/or” should be construed in the same fashion, i.e.,“one or more” of the elements so conjoined. Other elements may optionally be present other than the elements specifically identified by the“and/or” clause, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example, a reference to“A and/or B”, when used in conjunction with open-ended language such as“comprising” can refer, in one embodiment, to A only (optionally including elements other than B); in another embodiment, to B only (optionally including elements other than A); in yet another embodiment, to both A and B (optionally including other elements); etc.

As used herein in the specification and in the claims,“or” should be understood to have the same meaning as“and/or” as defined above. For example, when separating items in a list, “or” or“and/or” shall be interpreted as being inclusive, i.e., the inclusion of at least one, but also including more than one, of a number or list of elements, and, optionally, additional unlisted items. Only terms clearly indicated to the contrary, such as“only one of’ or“exactly one of,” or, when used in the claims,“consisting of,” will refer to the inclusion of exactly one element of a number or list of elements. In general, the term“or” as used herein shall only be interpreted as indicating exclusive alternatives (i.e.“one or the other but not both”) when preceded by terms of exclusivity, such as“either,”“one of,”“only one of,” or“exactly one of.”“Consisting essentially of,” when used in the claims, shall have its ordinary meaning as used in the field of patent law.

As used herein in the specification and in the claims, the phrase“at least one,” in reference to a list of one or more elements, should be understood to mean at least one element selected from any one or more of the elements in the list of elements, but not necessarily including at least one of each and every element specifically listed within the list of elements and not excluding any combinations of elements in the list of elements. This definition also allows that elements may optionally be present other than the elements specifically identified within the list of elements to which the phrase“at least one” refers, whether related or unrelated to those elements specifically identified. Thus, as a non-limiting example,“at least one of A and B” (or, equivalently,“at least one of A or B,” or, equivalently“at least one of A and/or B”) can refer, in one embodiment, to at least one, optionally including more than one, A, with no B present (and optionally including elements other than B); in another embodiment, to at least one, optionally including more than one, B, with no A present (and optionally including elements other than A); in yet another embodiment, to at least one, optionally including more than one, A, and at least one, optionally including more than one, B (and optionally including other elements); etc.

It should also be understood that, unless clearly indicated to the contrary, in any methods claimed herein that include more than one step or act, the order of the steps or acts of the method is not necessarily limited to the order in which the steps or acts of the method are recited.

In the claims, as well as in the specification above, all transitional phrases such as “comprising,”“including,”“carrying,”“having, “containing,”“involving,”“holding,” “composed of,” and the like are to be understood to be open-ended, i.e., to mean including but not limited to. Only the transitional phrases“consisting of’ and“consisting essentially of’ shall be closed or semi-closed transitional phrases, respectively, as set forth in the United States Patent Office Manual of Patent Examining Procedures, Section 2111.03.