Epigenetic priming enhances antitumor immunity in platinum-resistant ovarian cancer

Siqi Chen, Ping Xie, Matthew Cowan, Hao Huang, Horacio Cardenas, Russell Keathley, Edward J Tanner, Gini F Fleming, John W Moroney, Alok Pant, Azza M Akasha, Ramana V Davuluri, Masha Kocherginsky, Bin Zhang, Daniela Matei, Siqi Chen, Ping Xie, Matthew Cowan, Hao Huang, Horacio Cardenas, Russell Keathley, Edward J Tanner, Gini F Fleming, John W Moroney, Alok Pant, Azza M Akasha, Ramana V Davuluri, Masha Kocherginsky, Bin Zhang, Daniela Matei

Abstract

BackgroundImmune checkpoint inhibitors (ICIs) have modest activity in ovarian cancer (OC). To augment their activity, we used priming with the hypomethylating agent guadecitabine in a phase II study.MethodsEligible patients had platinum-resistant OC, normal organ function, measurable disease, and received up to 5 prior regimens. The treatment included guadecitabine (30 mg/m2) on days 1-4, and pembrolizumab (200 mg i.v.) on day 5, every 21 days. The primary endpoint was the response rate. Tumor biopsies, plasma, and PBMCs were obtained at baseline and after treatment.ResultsAmong 35 evaluable patients, 3 patients had partial responses (8.6%), and 8 (22.9%) patients had stable disease, resulting in a clinical benefit rate of 31.4% (95% CI: 16.9%-49.3%). The median duration of clinical benefit was 6.8 months. Long-interspersed element 1 (LINE1) was hypomethylated in post-treatment PBMCs, and methylomic and transcriptomic analyses showed activation of antitumor immunity in post-treatment biopsies. High-dimensional immune profiling of PBMCs showed a higher frequency of naive and/or central memory CD4+ T cells and of classical monocytes in patients with a durable clinical benefit or response (CBR). A higher baseline density of CD8+ T cells and CD20+ B cells and the presence of tertiary lymphoid structures in tumors were associated with a durable CBR.ConclusionEpigenetic priming using a hypomethylating agent with an ICI was feasible and resulted in a durable clinical benefit associated with immune responses in selected patients with recurrent OC.Trial registrationClinicalTrials.gov NCT02901899.FundingUS Army Medical Research and Material Command/Congressionally Directed Medical Research Programs (USAMRMC/CDMRP) grant W81XWH-17-0141; the Diana Princess of Wales Endowed Professorship and LCCTRAC funds from the Robert H. Lurie Comprehensive Cancer Center; Walter S. and Lucienne Driskill Immunotherapy Research funds; Astex Pharmaceuticals; Merck & Co.; National Cancer Institute (NCI), NIH grants CCSG P30 CA060553, CCSG P30 CA060553, and CA060553.

Keywords: Cancer; Cancer immunotherapy; Epigenetics; Immunology; Oncology.

Figures

Figure 1. G+P in recurrent OC.
Figure 1. G+P in recurrent OC.
(A) CONSORT diagram. CBR, clinical benefit or response. (B) Kaplan-Meier estimates of PFS (n = 43). (C) Duration of clinical benefit or response (months). PD, progressive disease. (D) LINE-1 methylation in PBMCs before G+P treatment (C1D1), after G+P treatment (C1D5), and 30 days after treatment discontinuation (D30). Data indicate the mean ± standard deviation. P values were determined with a mixed-effects model and Tukey’s multiple-comparison test. n = 34 pairs for C1D1 versus C1D5; n = 16 pairs for C1D1 versus D30. (E) Average methylation of CpGs (β values) measured using Epic arrays in C2D8 versus C1D1 tumor biopsies. Data indicate the mean ± standard deviation. P value was determined by paired t test. n = 11 pairs. (F) Volcano plot of DNA methylation (β values) in C2D8 versus C1D1 tumor biopsies (n = 11 pairs). adj, adjusted. (G) Top 10 pathways identified by WikiPathways enrichment analysis using 767 genes containing DM CpGs associated with CpG islands located in the gene body or promoter-associated region TSS200+TSS1500 (Illumina nomenclature) in C2D8 versus C1D1 tumor biopsies (n = 11 pairs).
Figure 2. Transcriptomic changes induced by G+P…
Figure 2. Transcriptomic changes induced by G+P treatment in recurrent OC.
(A) Volcano plot shows DEGs in post-treatment (C2D8) versus pretreatment (C1D1) tumor biopsies (n = 9 pairs). (B) Hierarchical clustering and heatmap show the 300 most variable genes between pre- and post-treatment biopsies. (C) Top 10 enriched biological pathways determined by GSEA of DEGs between pre- and post-treatment biopsies. (D) GSEA enrichment plots for the following biological pathways: “interactions between immune cells and microRNAs in tumor microenvironment,” “cancer immunotherapy by the PD1 blockade,” and “T cell antigen receptor TCR signaling pathway.” NES, normalized enrichment score. (E) Heatmap showing mRNA expression levels of the top 50 up- and downregulated genes (FDR <0.10) in pretreatment tumors from patients categorized as responders (durable CBR, n = 9) versus nonresponders (n = 7).
Figure 3. Mass spectrometric identification of differences…
Figure 3. Mass spectrometric identification of differences in subsets of PBMCs from patients with a durable CBR and nonresponders before treatment with G+P.
(A) Exemplified tSNE visualization of overlaid cell population composition in PBMCs from an initial cohort of nonresponders (n = 6) and durable CBR patients (n = 4) before the initiation of therapy (C1D1). (B) SPADE analysis of total immune cell populations in PBMCs from nonresponders (upper left) and durable CBR patients (upper right); as well as the subsets among total CD4+ T cells from nonresponders (lower left, n = 6) and durable CBR patients (lower right, n = 4) at C1D1. The size and color of each node correspond to the number of cells. (C) Left: CITRUS analysis showing a visual representation of unsupervised hierarchical clustering of CD4 cells and visualization of the clusters that were part of the significant results (in red). Right: Abundance plots for the significant cluster 2478 (of CD4 cells) in nonresponders (NR) and responders (CBR). (D) Heatmap represents the median expression levels of indicated markers within the cluster 2478 from nonresponders and responders (CBR).
Figure 4. Differences in the frequencies of…
Figure 4. Differences in the frequencies of myeloid cell populations between patients with a durable CBR and nonresponders before treatment with G+P, by spectral cytometry.
(A) Exemplified tSNE visualization of the overlaid cell population composition in PBMCs from an extended cohort of nonresponders (n = 9) and patients with durable CBRs (n = 6) before the initiation of therapy (C1D1). (B) edgeR analysis identified myeloid cellular populations with significant differences in relative abundance between nonresponders and patients with a durable CBR. (C) Differences in the percentages of NC monocytes, DCs, and classical monocytes among live PBMCs from nonresponders or durable CBR patients at C1D1. Data indicate the mean ± standard deviation. *P < 0.05 and **P < 0.01, by 2-tailed t test with multiple-comparison correction using the Benjamini-Hochberg adjustment. (D) Heatmap represents the median expression for markers (CD279, CD274, and CD38) that were differentially expressed (adjusted P < 0.05) in NC monocytes, DCs, and classical monocytes between nonresponders and durable CBR patients.
Figure 5. Identification of metaclusters in PBMCs…
Figure 5. Identification of metaclusters in PBMCs with a significant difference between patients with a durable CBR and nonresponders before treatment with G+P.
(A) Exemplified tSNE visualization of overlaid unsupervised metaclusters in PBMCs using the FlowSOM algorithm from an extended cohort of nonresponders (n = 9) and patients with a durable CBR (n = 6) at C1D1, prior to therapy. (B) edgeR analysis identified metaclusters with significant differences in relative abundance between nonresponders and patients with a durable CBR. (C) Differences in the percentages of unsupervised metaclusters in PBMCs from nonresponders and patients with a durable CBR at C1D1. Data indicate the mean ± standard deviation. *P < 0.05 and **P < 0.01, by 2-tailed t test with multiple-comparison correction using the Benjamini-Hochberg adjustment. (D) Heatmap represents the median expression levels of the indicated markers within the metaclusters that had significant differences in relative abundance between nonresponders and patients with a durable CBR.
Figure 6. Distinct features of TILs between…
Figure 6. Distinct features of TILs between nonresponders and patients with a durable CBR.
(A) Representative multiplex image (upper left panel) with inset (upper right) of cytotoxic T cells (CD3+CD8+), B cells (CD20+), macrophages (CD68+), and Tregs (CD3+CD8–FOXP3+) in a responder patient after treatment with G+P, measured by mIHC. (B and C) Density of tumor-infiltrating CD8+ T cells (B) and CD20+ B cells (C) in the compartments of the tumor nest and stroma from patients with a durable CBR (n = 5) and nonresponders (n = 9). (DF) Spatial characterization among TILs and tumor cells from patients with a durable CBR (n = 5) and nonresponders (n = 9). (D) Distance from TILs (red) to tumor cells (blue). (E) Touching events between TILs (yellow) and tumor cells (red). (F) Touching events among TILs. (G) Representative image of mIHC staining of putative TLSs in a patient with a durable CBR after treatment with G+P. (H) Comparison of putative TLSs at C1D1 (left) and C2D8 (right) between patients with a durable CBR (n = 5) and nonresponders (n = 9). Box and whiskers represent the mean ± standard deviation, and each dot represents 1 patient. Original magnification, ×40 (A and DG). Higher-magnification images in and A and G were generated in Photoshop by selecting the indicated areas using the crop tool, and then expanding the areas. *P < 0.05, **P < 0.01, and ***P < 0.001, by 2-way ANOVA followed by multiple-comparison correction (BF) and the Mann-Whitney U test (H).
Figure 7. Increased expression levels of PD-L1…
Figure 7. Increased expression levels of PD-L1 and A2AR within tumors related to a favorable clinical response to G+P treatment.
(A) Representative multiplex (upper panel) and singlet immunostaining (lower panels) images for the following markers: PD-L1, NY-ESO-1, A2AR, CD8, PanCK, and DAPI. Original magnification, ×40. (B) Comparison of PD-L1 expression in tumors by combined positive score (CPS) between patients with a durable CBR (n = 5) and nonresponders (n = 9). (C and D) Expression levels of PD-L1 on total cells (C) and on PanCK+ tumor cells (D) at C1D1 and C2D8 between patients with a durable CBR (n = 5) and nonresponders (n = 9). (E) Touching events between PD-L1+ cells and CD8+ cells at C1D1 and C2D8 in cells from patients with a durable CBR (n = 5) and nonresponders (n = 9). (F and G) Expression levels of A2AR on tumor cells (F) or on tumor nest–infiltrating CD8+ T cells (G) at C1D1 and C2D8 between patients with a durable CBR (n = 5) and nonresponders (n = 9). Box and whiskers represent the mean ± standard deviation, and each point represents 1 patient. *P < 0.05, **P < 0.01, and ***P < 0.001, by 2-way ANOVA with multiple-comparison correction (B and EG), and 2-tailed paired t test (C and D).

References

    1. Garon EB, et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N Engl J Med. 2015;372(21):2018–2028. doi: 10.1056/NEJMoa1501824.
    1. Barber E, Matei D. Immunotherapy in ovarian cancer: we are not there yet. Lancet Oncol. 2021;22(7):903–905. doi: 10.1016/S1470-2045(21)00303-X.
    1. Varga A, et al. Pembrolizumab in patients with programmed death ligand 1-positive advanced ovarian cancer: analysis of KEYNOTE-028. Gynecol Oncol. 2019;152(2):243–250. doi: 10.1016/j.ygyno.2018.11.017.
    1. Disis ML, et al. Efficacy and safety of avelumab for patients with recurrent or refractory ovarian cancer: phase 1b results from the JAVELIN Solid Tumor Trial. JAMA Oncol. 2019;5(3):393–401. doi: 10.1001/jamaoncol.2018.6258.
    1. Pujade-Lauraine E, et al. Avelumab alone or in combination with chemotherapy versus chemotherapy alone in platinum-resistant or platinum-refractory ovarian cancer (JAVELIN Ovarian 200): an open-label, three-arm, randomised, phase 3 study. Lancet Oncol. 2021;22(7):1034–1046. doi: 10.1016/S1470-2045(21)00216-3.
    1. Jones PA, Baylin SB. The fundamental role of epigenetic events in cancer. Nat Rev Genet. 2002;3(6):415–428. doi: 10.1038/nrg816.
    1. Chiappinelli KB, et al. Inhibiting DNA Methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses. Cell. 2015;162(5):974–986. doi: 10.1016/j.cell.2015.07.011.
    1. Roulois D, et al. DNA-demethylating agents target colorectal cancer cells by inducing viral mimicry by endogenous transcripts. Cell. 2015;162(5):961–973. doi: 10.1016/j.cell.2015.07.056.
    1. Yang H, et al. Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents. Leukemia. 2014;28(6):1280–1288. doi: 10.1038/leu.2013.355.
    1. Chiappinelli KB, et al. Combining epigenetic and immunotherapy to combat cancer. Cancer Res. 2016;76(7):1683–1689. doi: 10.1158/0008-5472.CAN-15-2125.
    1. Stone ML, et al. Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden. Proc Natl Acad Sci U S A. 2017;114(51):E10981–E10990.
    1. Balch C, et al. New anti-cancer strategies: epigenetic therapies and biomarkers. Front Biosci. 2005;10:1897–1931. doi: 10.2741/1668.
    1. Adair SJ, Hogan KT. Treatment of ovarian cancer cell lines with 5-aza-2’-deoxycytidine upregulates the expression of cancer-testis antigens and class I major histocompatibility complex-encoded molecules. Cancer Immunol Immunother. 2009;58(4):589–601. doi: 10.1007/s00262-008-0582-6.
    1. Woloszynska-Read A, et al. Intertumor and intratumor NY-ESO-1 expression heterogeneity is associated with promoter-specific and global DNA methylation status in ovarian cancer. Clin Cancer Res. 2008;14(11):3283–3290. doi: 10.1158/1078-0432.CCR-07-5279.
    1. Odunsi K, et al. Epigenetic potentiation of NY-ESO-1 vaccine therapy in human ovarian cancer. Cancer Immunol Res. 2014;2(1):37–49. doi: 10.1158/2326-6066.CIR-13-0126.
    1. Peng D, et al. Epigenetic silencing of TH1-type chemokines shapes tumour immunity and immunotherapy. Nature. 2015;527(7577):249–253. doi: 10.1038/nature15520.
    1. Fang F, et al. Genomic and epigenomic signatures in ovarian cancer associated with resensitization to platinum drugs. Cancer Res. 2018;78(3):631–644. doi: 10.1158/0008-5472.CAN-17-1492.
    1. Matei D, et al. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res. 2012;72(9):2197–2205. doi: 10.1158/0008-5472.CAN-11-3909.
    1. Yang AS, et al. A simple method for estimating global DNA methylation using bisulfite PCR of repetitive DNA elements. Nucleic Acids Res. 2004;32(3):e38. doi: 10.1093/nar/gnh032.
    1. Weisenberger DJ, et al. Analysis of repetitive element DNA methylation by MethyLight. Nucleic Acids Res. 2005;33(21):6823–6836. doi: 10.1093/nar/gki987.
    1. Bendall ML, et al. Telescope: characterization of the retrotranscriptome by accurate estimation of transposable element expression. PLoS Comput Biol. 2019;15(9):e1006453. doi: 10.1371/journal.pcbi.1006453.
    1. McDonald JI, et al. Epigenetic therapies in ovarian cancer alter repetitive element expression in a TP53-dependent manner. Cancer Res. 2021;81(20):5176–5189. doi: 10.1158/0008-5472.CAN-20-4243.
    1. Laino AS, et al. Serum interleukin-6 and C-reactive protein are associated with survival in melanoma patients receiving immune checkpoint inhibition. J Immunother Cancer. 2020;8(1):e000842. doi: 10.1136/jitc-2020-000842.
    1. Schalper KA, et al. Elevated serum interleukin-8 is associated with enhanced intratumor neutrophils and reduced clinical benefit of immune-checkpoint inhibitors. Nat Med. 2020;26(5):688–692. doi: 10.1038/s41591-020-0856-x.
    1. Tsukamoto H, et al. Combined blockade of IL6 and PD-1/PD-L1 signaling abrogates mutual regulation of their immunosuppressive effects in the tumor microenvironment. Cancer Res. 2018;78(17):5011–5022. doi: 10.1158/0008-5472.CAN-18-0118.
    1. Yuen KC, et al. High systemic and tumor-associated IL-8 correlates with reduced clinical benefit of PD-L1 blockade. Nat Med. 2020;26(5):693–8. doi: 10.1038/s41591-020-0860-1.
    1. Issa JP, et al. Safety and tolerability of guadecitabine (SGI-110) in patients with myelodysplastic syndrome and acute myeloid leukaemia: a multicentre, randomised, dose-escalation phase 1 study. Lancet Oncol. 2015;16(9):1099–1110. doi: 10.1016/S1470-2045(15)00038-8.
    1. Kantarjian HM, et al. Guadecitabine (SGI-110) in treatment-naive patients with acute myeloid leukaemia: phase 2 results from a multicentre, randomised, phase 1/2 trial. Lancet Oncol. 2017;18(10):1317–1326. doi: 10.1016/S1470-2045(17)30576-4.
    1. Matei D, et al. A phase I clinical trial of guadecitabine and carboplatin in platinum-resistant, recurrent ovarian cancer: clinical, pharmacokinetic, and pharmacodynamic analyses. Clin Cancer Res. 2018;24(10):2285–2293. doi: 10.1158/1078-0432.CCR-17-3055.
    1. Oza AM, et al. A randomized phase II trial of epigenetic priming with guadecitabine and carboplatin in platinum-resistant, recurrent ovarian cancer. Clin Cancer Res. 2020;26(5):1009–1016. doi: 10.1158/1078-0432.CCR-19-1638.
    1. Fang F, et al. Decitabine reactivated pathways in platinum resistant ovarian cancer. Oncotarget. 2014;5(11):3579–3589. doi: 10.18632/oncotarget.1961.
    1. Li M, et al. Integrated analysis of DNA methylation and gene expression reveals specific signaling pathways associated with platinum resistance in ovarian cancer. BMC Med Genomics. 2009;2:34. doi: 10.1186/1755-8794-2-34.
    1. Noonepalle SK, et al. Promoter methylation modulates indoleamine 2,3-dioxygenase 1 induction by activated T cells in human breast cancers. Cancer Immunol Res. 2017;5(4):330–344. doi: 10.1158/2326-6066.CIR-16-0182.
    1. Xue ZT, et al. An epigenetic mechanism for high, synergistic expression of indoleamine 2,3-dioxygenase 1 (IDO1) by combined treatment with zebularine and IFN-γ: potential therapeutic use in autoimmune diseases. Mol Immunol. 2012;51(2):101–111. doi: 10.1016/j.molimm.2012.01.006.
    1. Klumper N, et al. LAG3 (LAG-3, CD223) DNA methylation correlates with LAG3 expression by tumor and immune cells, immune cell infiltration, and overall survival in clear cell renal cell carcinoma. J Immunother Cancer. 2020;8(1):undefined.
    1. Kagamu H, et al. CD4+ T-cell immunity in the peripheral blood correlates with response to anti-PD-1 therapy. Cancer Immunol Res. 2020;8(3):334–344. doi: 10.1158/2326-6066.CIR-19-0574.
    1. Takeuchi Y, et al. Clinical response to PD-1 blockade correlates with a sub-fraction of peripheral central memory CD4+ T cells in patients with malignant melanoma. Int Immunol. 2018;30(1):13–22. doi: 10.1093/intimm/dxx073.
    1. Zuazo M, et al. Functional systemic CD4 immunity is required for clinical responses to PD-L1/PD-1 blockade therapy. EMBO Mol Med. 2019;11(7):e10293.
    1. Krieg C, et al. Author Correction: High-dimensional single-cell analysis predicts response to anti-PD-1 immunotherapy. Nat Med. 2018;24(11):1773–1775. doi: 10.1038/s41591-018-0094-7.
    1. Guo FF, Cui JW. The role of tumor-infiltrating B cells in tumor immunity. J Oncol. 2019;2019:2592419.
    1. Cabrita R, et al. Tertiary lymphoid structures improve immunotherapy and survival in melanoma. Nature. 2020;577(7791):561–5. doi: 10.1038/s41586-019-1914-8.
    1. Helmink BA, et al. B cells and tertiary lymphoid structures promote immunotherapy response. Nature. 2020;577(7791):549–555. doi: 10.1038/s41586-019-1922-8.
    1. Petitprez F, et al. B cells are associated with survival and immunotherapy response in sarcoma. Nature. 2020;577(7791):556–560. doi: 10.1038/s41586-019-1906-8.
    1. Lauss M, et al. B cells and tertiary lymphoid structures: friends or foes in cancer immunotherapy? Clin Cancer Res. 2021;28(9):1751–1758.
    1. Meylan M, et al. Tertiary lymphoid structures generate and propagate anti-tumor antibody-producing plasma cells in renal cell cancer. Immunity. 2022;55(3):527–541. doi: 10.1016/j.immuni.2022.02.001.
    1. Zaretsky JM, et al. Mutations associated with acquired resistance to PD-1 blockade in melanoma. N Engl J Med. 2016;375(9):819–829. doi: 10.1056/NEJMoa1604958.
    1. Jin D, et al. CD73 on tumor cells impairs antitumor T-cell responses: a novel mechanism of tumor-induced immune suppression. Cancer Res. 2010;70(6):2245–2255. doi: 10.1158/0008-5472.CAN-09-3109.
    1. Ohta A, et al. A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci U S A. 2006;103(35):13132–13137. doi: 10.1073/pnas.0605251103.
    1. Beavis PA, et al. Adenosine receptor 2A blockade increases the efficacy of anti-PD-1 through enhanced antitumor T-cell responses. Cancer Immunol Res. 2015;3(5):506–517. doi: 10.1158/2326-6066.CIR-14-0211.
    1. Fong L, et al. Adenosine 2A receptor blockade as an immunotherapy for treatment-refractory renal cell cancer. Cancer Discov. 2020;10(1):40–53. doi: 10.1158/-19-0980.
    1. Willingham SB, et al. A2AR antagonism with CPI-444 induces antitumor responses and augments efficacy to anti-PD-(L)1 and anti-CTLA-4 in preclinical models. Cancer Immunol Res. 2018;6(10):1136–1149. doi: 10.1158/2326-6066.CIR-18-0056.
    1. Falconi A, et al. On the role of adenosine A2A receptor gene transcriptional regulation in Parkinson’s Disease. Front Neurosci. 2019;13:683. doi: 10.3389/fnins.2019.00683.
    1. Vogt TJ, et al. Detailed analysis of adenosine A2a receptor (ADORA2A) and CD73 (5’-nucleotidase, ecto, NT5E) methylation and gene expression in head and neck squamous cell carcinoma patients. Oncoimmunology. 2018;7(8):e1452579.
    1. Cardenas H, et al. TGF-β induces global changes in DNA methylation during the epithelial-to-mesenchymal transition in ovarian cancer cells. Epigenetics. 2014;9(11):1461–1472. doi: 10.4161/15592294.2014.971608.
    1. Bendall SC, et al. Single-cell mass cytometry of differential immune and drug responses across a human hematopoietic continuum. Science. 2011;332(6030):687–696. doi: 10.1126/science.1198704.
    1. Wei SC, et al. Distinct cellular mechanisms underlie anti-CTLA-4 and Anti-PD-1 Checkpoint Blockade. Cell. 2017;170(6):1120–1133. doi: 10.1016/j.cell.2017.07.024.
    1. Amir el AD, et al. viSNE enables visualization of high dimensional single-cell data and reveals phenotypic heterogeneity of leukemia. Nat Biotechnol. 2013;31(6):545–552. doi: 10.1038/nbt.2594.
    1. Van Gassen S, et al. FlowSOM: u+sing self-organizing maps for visualization and interpretation of cytometry data. Cytometry A. 2015;87(7):636–645. doi: 10.1002/cyto.a.22625.
    1. Qiu P, et al. Extracting a cellular hierarchy from high-dimensional cytometry data with SPADE. Nat Biotechnol. 2011;29(10):886–891. doi: 10.1038/nbt.1991.
    1. Bruggner RV, et al. Automated identification of stratifying signatures in cellular subpopulations. Proc Natl Acad Sci U S A. 2014;111(26):E2770–E2777.
    1. Tusher VG, et al. Significance analysis of microarrays applied to the ionizing radiation response. Proc Natl Acad Sci U S A. 2001;98(9):5116–5121. doi: 10.1073/pnas.091062498.
    1. Robinson MD, et al. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics. 2010;26(1):139–140. doi: 10.1093/bioinformatics/btp616.
    1. Poropatich K, et al. OX40+ plasmacytoid dendritic cells in the tumor microenvironment promote antitumor immunity. J Clin Invest. 2020;130(7):3528–3542. doi: 10.1172/JCI131992.
    1. Benjamini Y, Yekutieli, D The control of the false discovery rate in multiple testing under dependency. Annal Stat. 2001;29(4):1165–1188. doi: 10.1214/aos/1013699998.

Source: PubMed

3
Sottoscrivi