Please wait a minute...
Frontiers of Medicine

ISSN 2095-0217

ISSN 2095-0225(Online)

CN 11-5983/R

Postal Subscription Code 80-967

2018 Impact Factor: 1.847

Front. Med.    2024, Vol. 18 Issue (2) : 357-374    https://doi.org/10.1007/s11684-023-1016-8
Decitabine induces IRF7-mediated immune responses in p53-mutated triple-negative breast cancer: a clinical and translational study
Haoyu Wang1, Zhengyuan Wang2, Zheng Wang1, Xiaoyang Li3, Yuntong Li2, Ni Yan2, Lili Wu2, Ying Liang2, Jiale Wu2, Huaxin Song2, Qing Qu1, Jiahui Huang1, Chunkang Chang4, Kunwei Shen1(), Xiaosong Chen1(), Min Lu2()
1. Department of General Surgery, Comprehensive Breast Health Center, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
2. Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine (Shanghai), Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
3. Department of Hematology, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
4. Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People’s Hospital, Shanghai 200025, China
 Download: PDF(3834 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

p53 is mutated in half of cancer cases. However, no p53-targeting drugs have been approved. Here, we reposition decitabine for triple-negative breast cancer (TNBC), a subtype with frequent p53 mutations and extremely poor prognosis. In a retrospective study on tissue microarrays with 132 TNBC cases, DNMT1 overexpression was associated with p53 mutations (P = 0.037) and poor overall survival (OS) (P = 0.010). In a prospective DEciTabinE and Carboplatin in TNBC (DETECT) trial (NCT03295552), decitabine with carboplatin produced an objective response rate (ORR) of 42% in 12 patients with stage IV TNBC. Among the 9 trialed patients with available TP53 sequencing results, the 6 patients with p53 mutations had higher ORR (3/6 vs. 0/3) and better OS (16.0 vs. 4.0 months) than the patients with wild-type p53. In a mechanistic study, isogenic TNBC cell lines harboring DETECT-derived p53 mutations exhibited higher DNMT1 expression and decitabine sensitivity than the cell line with wild-type p53. In the DETECT trial, decitabine induced strong immune responses featuring the striking upregulation of the innate immune player IRF7 in the p53-mutated TNBC cell line (upregulation by 16-fold) and the most responsive patient with TNBC. Our integrative studies reveal the potential of repurposing decitabine for the treatment of p53-mutated TNBC and suggest IRF7 as a potential biomarker for decitabine-based treatments.

Keywords p53 mutation      triple-negative breast cancer      decitabine      DNMT1      IRF7      innate immune response     
Corresponding Author(s): Kunwei Shen,Xiaosong Chen,Min Lu   
Just Accepted Date: 20 October 2023   Online First Date: 25 December 2023    Issue Date: 27 May 2024
 Cite this article:   
Haoyu Wang,Zhengyuan Wang,Zheng Wang, et al. Decitabine induces IRF7-mediated immune responses in p53-mutated triple-negative breast cancer: a clinical and translational study[J]. Front. Med., 2024, 18(2): 357-374.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-023-1016-8
https://academic.hep.com.cn/fmd/EN/Y2024/V18/I2/357
Fig.1  DNMT1 overexpression was associated with p53 mutation and poor prognosis in TNBC. (A) Integrated heatmap of biomarkers (IHC of p53, DNMT1, DNMT3A, DNMT3B, and 5mC), clinicopathological features and prognosis in the TMA cohort consisting of 132 patients with stage I–III TNBC. (B) Box plots of the IHC scores of DNMT1, DNMT3A, DNMT3B, and 5mC in tumors with different p53 IHC patterns. WT, wild-type p53 staining pattern; MUT, mutant p53 staining pattern. (C) Representative IHC of adjacent sections showing scattered, absent, or diffused p53 and DNMT1 staining patterns. (D) Kaplan–Meier plots of RFS and OS stratified by DNMT1 expression levels. HR, hazard ratio; CI, confidence interval. (E) Forest plot of multivariate regression models predicting RFS and OS by combining biomarkers and clinicopathological features. Statistically significant features are marked in red. (F) Kaplan–Meier plots of RFS and OS for p53-mutated patients stratified by DNMT1 expression levels. (G) Forest plot of multivariate regression models predicting RFS and OS by combining biomarkers and clinicopathological features in p53-mutated patients. Statistically significant features are marked in red.
Fig.2  Response and AEs in the DETECT trial. (A) Eligibility, treatment design, and endpoints of the DETECT trial. (B) Swimmer plot illustrating the best response, PFS (progression-free survival), postprogression survival (PPS), and survival status of each patient. PD, progressed disease; SD, stable disease; PR, partial response. (C) Waterfall plot illustrating the best response of the 12 trialed patients during treatment. (D) Spider plot demonstrating the size changes in the targeted lesions from the baseline every 2 treatment cycles. (E) Representative radiological images showing changes in the targeted lesions of patients #10 and #6. Targeted lesions are marked with red circles. Pt, patient. (F and G) Kaplan–Meier plots of the PFS and OS of patients enrolled in DETECT. The median survival time is shown. (H) Observed hematological and nonhematological AEs and number of treatment cycles for the 12 trialed patients. Upper panel showing the observed hematological and nonhematological AEs. The number of treatment cycles for the 12 trialed patients is shown in the lower panel.
Characteristics N %
Age, years
Median 53
Range 35–68
< 45 3 25
45–55 5 42
> 55 4 33
ECOG performance status
0 9 75
1 3 25
BCFI, months
Median 11
Range 4–50
Neoadjuvant therapy
No 8 67
Yes 3 25
NA 1 8
Stage at initial diagnosis
I 2 17
II 4 33
III 5 42
IV 1 8
Adjuvant chemotherapy
Anthracycline and taxane 9 75
Capecitabine 2 17
NA 1 8
Site of metastasis
Lung 5 42
Liver 1 8
Lymph nodes 5 42
Breast 1 8
Bone 2 17
Previous lines of therapy
0 9 75
1 3 25
Capecitabine 2 17
Capecitabine + vinorelbine 1 8
Tab.1  Demographics and baseline characteristics of patients enrolled in the DETECT trial
AE Grades 1 and 2 Grade 3 Grade 4
Neutropenia 5 (41.7%) 5 (41.7%) 2 (16.7%)
Leukopenia 9 (75%) 2 (16.7%) 1 (8.3%)
Anemia 6 (50.0%) 0 0
Thrombocytopenia 6 (50.0%) 4 (33.3%) 1 (8.3%)
Fatigue 7 (58.3%) 0 0
Nausea 12 (100%) 0 0
Vomiting 8 (66.7%) 0 0
Constipation 4 (33.3%) 0 0
Fever 2 (16.7%) 0 0
AST elevation 4 (33.3%) 0 0
ALT elevation 4 (33.3%) 0 0
Tab.2  Summary of AEs
Fig.3  Correlation between p53 mutations and responses in the DETECT trial. (A) DNA demethylation of PBMCs from Pt #10, #11, and #12. Pt, patient. PBMCs were collected and gDNA was extracted. The representative LINE-1 methylation levels were determined. The right panel shows the quantified LINE-1 methylation levels. (B) Location of the 6 p53 mutations identified in the DETECT trial. (C) Swimmer plot illustrating the best response, PFS, PPS, and survival status of patients with different p53 mutation statuses and DNMT1 expression levels. (D) Waterfall plot illustrating the best response among the trialed patients with different p53 mutation statuses. Wild-type p53 is marked in blue, mutant p53 in orange, and primary tumor tissues unavailable for sequencing are in gray. (E) Spider plot demonstrating the size changes of the targeted lesions with different p53 mutation statuses from the baseline at every 2 treatment cycles. Wild-type p53 is marked in blue, mutant p53 is in orange, and primary tumor tissues unavailable for sequencing are in gray. (F and G) Kaplan–Meier plots showing PFS and OS stratified by p53 mutation status. The median survival time of each subgroup is shown. (H) Waterfall plot illustrating the best response in the trialed patients with different DNMT1 expression levels. High DNMT1 expression is marked in dark green, low DNMT1 expression is marked in light green, and primary tumor tissues unavailable for sequencing are in gray. (I) Spider plot demonstrating the size changes in the targeted lesions with different DNMT1 expression levels from the baseline at every 2 treatment cycles. High DNMT1 high expression is marked in dark green, low DNMT1 expression is marked in light green, and primary tumor tissues unavailable for sequencing are in gray. (J and K) Kaplan–Meier plots showing PFS and OS stratified by different DNMT1 expression levels. The median survival time of each subgroup is shown.
Fig.4  Decitabine preferentially inhibited isogenic TNBC cell lines harboring DETECT-derived p53 mutations. (A) Left panel showing the structural view of the DBD of wild-type p53 (3KMD, chain A) with the 4 residues of interest and 2 representative DNA-contacting residues shown as yellow and brown sticks, respectively. The solvent-accessible surface area of the indicated residues was calculated by PyMOL and shown. Right panel, the interaction networks of the 4 residues of interest with the involved hydrogen bonds shown as dotted lines. (B) Luciferase reporter assay indicating the transcriptional activity of the indicated p53 mutants on CDKN1A in p53-null H1299 cells. (C) Immunoblotting of DNMT1, DNMT3A, and DNMT3B in the TNBC cell line HCC1937 after treatment with a concentration gradient of decitabine. (D) Relative mRNA levels of DNMT1 in isogenic HCC1937 cell lines infected with wild-type p53 and DETECT-derived p53 mutants. (E) Protein levels of DNMT1 in isogenic HCC1937 cell lines expressing DETECT-derived p53 mutations. p21 was used as the control for the functional wild-type p53. (F) Luciferase reporter assay on the transcriptional activity of wild-type p53 on DNMT1 and CDKN1A promoters in p53-null H1299 cells. (G) Relative mRNA levels of DNMT1 and CDKN1A in p53-null HCC1937 cells transfected with the vector, wild-type p53, p53-L25Q/W26S (p5325,26), or p53-L25Q/W26S/F53Q/F54S (p5325,26,53,54). (H) Immunoblotting of DNMT1 and CDKN1A in HCC1937 cells transfected with the vector, wild-type p53, p5325,26, or p5325,26,53,54. (I) Isogenic HCC1937 cell lines were treated with different doses of decitabine every 24 h for 5 days then subjected to cell viability determination. (J) HCC1937 and BT549 cell lines were transfected with nontarget RNAi or DNMT1 RNAi. The protein levels of DNMT1 were then determined. (K) HCC1937 and BT549 cell lines transfected with nontarget RNAi or DNMT1 RNAi were treated with different doses of decitabine for 5 days and then subjected to cell viability determination. HCC1937 is marked in yellow and BT549 is marked with purple bars.
Fig.5  Decitabine potently induced IRF7-mediated immune responses in p53-deficient TNBC. (A) mRNA was isolated from p53-R282W-harboring HCC1937 cells with or without 0.3 μM decitabine treatment for 5 days then subjected to RNA-seq. Dot plot showing the FPKM values of the transcriptomes of the indicated cells. Orange dots, upregulated DEGs (fold change > 2, Gfold > 0.3). Blue dots, downregulated DEGs (fold change < 0.5, Gfold < −0.3). Gray dots, other genes classified as not differentially expressed. (B and C) Enrichment assays based on the Gene Ontology biological process and REACTOME for the 870 decitabine-upregulated DEGs as in (A). DEG numbers for each term were marked with green dots. Asterisks represent items involved in immune response. expre., expression; signal., signaling. (D) Bar plots showing the ratio of upregulated DEGs in the indicated terms and the ratio of upregulated DEGs in the whole genome. (E) Heatmap showing the expression profiles of the 613 IRGs recorded in the ImmPort database in the indicated cells. The myeloid malignant cell line Thp-1 harboring p53-R282W was included for comparison. The genes of interest are labeled. (F) Dot plot showing the fold change of the mRNA levels of the 613 IRGs upon decitabine treatment in the indicated cell lines. The genes of interest were labeled. Orange dots, IRGs preferentially upregulated in HCC1937. Blue dots, IRGs preferentially upregulated in Thp-1. Black dots, IRGs regulated in 2 cell lines. Gray dots, other unclassified IRGs. (G) PPI network for the decitabine-upregulated IRGs in HCC1937 cells based on the STRING database with MCL algorithms. Right panel showing the cluster of the specific PPI network of interest. (H) Volcano plot of the correlations between the immune cell infiltration score determined by using the ssGSEA algorithm and the expression level of IRF7 mRNA in the TNBC cohort in TCGA. Yellow circles mark immune cell types with correlation coefficients of 0.3–0.5 and red circles mark those with coefficients > 0.5. (I) Scatter plots of immune cell infiltration scores for the indicated immune cell types against the mRNA level of IRF7 in the TCGA TNBC cohort as in (G). (J) Hematoxylin and eosin staining and IRF7 and CD8 IHC staining of tumor tissues from the patient with the best response (#10) before treatment (cycle 1 day 0) and after 6 cycles of treatment.
Fig.6  Summary of the study. Left panel, in the retrospective TMA study, DNMT1 overexpression was associated with p53 mutation and poor OS in TNBC. Middle panel, in the prospective DETECT trial on the combination of decitabine and carboplatin in 12 patients with metastatic stage IV TNBC, an ORR of 42% was achieved, wherein patients with p53-mutated TNBC preferentially benefited (ORR 3/6 vs. 0/3). Right panel, in the matched mechanistical study on isogenic TNBC cell lines, DETECT-derived p53 mutations conferred TNBC cells with sensitivity to decitabine treatment by upregulating DNMT1. Compared with the treatment for myeloid malignancy, decitabine potently induced innate immune response featuring striking IRF7 upregulation and consequent T cell activation in p53-mutated TNBC cell lines and the best patient response in DETECT trial. upreg., upregulation; pre-treat., pre-treatment; post-treat., post-treatment.
1 IF Tannock, JA Hickman. Limits to personalized cancer medicine. N Engl J Med 2016; 375(13): 1289–1294
https://doi.org/10.1056/NEJMsb1607705
2 V Prasad. Perspective: The precision-oncology illusion. Nature 2016; 537(7619): S63
https://doi.org/10.1038/537S63a
3 AC Joerger, AR Fersht. The p53 pathway: origins, inactivation in cancer, and emerging therapeutic approaches. Annu Rev Biochem 2016; 85(1): 375–404
https://doi.org/10.1146/annurev-biochem-060815-014710
4 PAJ Muller, KH Vousden. Mutant p53 in cancer: new functions and therapeutic opportunities. Cancer Cell 2014; 25(3): 304–317
https://doi.org/10.1016/j.ccr.2014.01.021
5 PAJ Muller, KH Vousden. p53 mutations in cancer. Nat Cell Biol 2013; 15(1): 2–8
https://doi.org/10.1038/ncb2641
6 K Sabapathy, DP Lane. Therapeutic targeting of p53: all mutants are equal, but some mutants are more equal than others. Nat Rev Clin Oncol 2018; 15(1): 13–30
https://doi.org/10.1038/nrclinonc.2017.151
7 SN Loh. Arsenic and an old place: rescuing p53 mutants in cancer. Cancer Cell 2021; 39(2): 140–142
https://doi.org/10.1016/j.ccell.2021.01.012
8 L Gummlich. ATO stabilizes structural p53 mutants. Nat Rev Cancer 2021; 21(3): 141
https://doi.org/10.1038/s41568-021-00337-1
9 S Chen, JL Wu, Y Liang, YG Tang, HX Song, LL Wu, YF Xing, N Yan, YT Li, ZY Wang, SJ Xiao, X Lu, SJ Chen, M Lu. Arsenic trioxide rescues structural p53 mutations through a cryptic allosteric site. Cancer Cell 2021; 39(2): 225–239.e8
https://doi.org/10.1016/j.ccell.2020.11.013
10 A Basu, NE Bodycombe, JH Cheah, EV Price, K Liu, GI Schaefer, RY Ebright, ML Stewart, D Ito, S Wang, AL Bracha, T Liefeld, M Wawer, JC Gilbert, AJ Wilson, N Stransky, GV Kryukov, V Dancik, J Barretina, LA Garraway, CSY Hon, B Munoz, JA Bittker, BR Stockwell, D Khabele, AM Stern, PA Clemons, AF Shamji, SL Schreiber. An interactive resource to identify cancer genetic and lineage dependencies targeted by small molecules. Cell 2013; 154(5): 1151–1161
https://doi.org/10.1016/j.cell.2013.08.003
11 S Leijen, RMJM van Geel, GS Sonke, D de Jong, EH Rosenberg, S Marchetti, D Pluim, E van Werkhoven, S Rose, MA Lee, T Freshwater, JH Beijnen, JHM Schellens. Phase II study of WEE1 inhibitor AZD1775 plus carboplatin in patients with TP53-mutated ovarian cancer refractory or resistant to first-line therapy within 3 months. J Clin Oncol 2016; 34(36): 4354–4361
https://doi.org/10.1200/JCO.2016.67.5942
12 L Xu, ZH Gu, Y Li, JL Zhang, CK Chang, CM Pan, JY Shi, Y Shen, B Chen, YY Wang, L Jiang, J Lu, X Xu, JL Tan, Y Chen, SY Wang, X Li, Z Chen, SJ Chen. Genomic landscape of CD34+ hematopoietic cells in myelodysplastic syndrome and gene mutation profiles as prognostic markers. Proc Natl Acad Sci U S A 2014; 111(23): 8589–8594
https://doi.org/10.1073/pnas.1407688111
13 C Chang, Y Zhao, F Xu, X Li. A primary study of the gene mutations in predicting treatment response to decitabine in patients with MDS. Blood 2015; 126(23): 1689
https://doi.org/10.1182/blood.V126.23.1689.1689
14 CK Chang, YS Zhao, F Xu, J Guo, Z Zhang, Q He, D Wu, LY Wu, JY Su, LX Song, C Xiao, X Li. TP53 mutations predict decitabine-induced complete responses in patients with myelodysplastic syndromes. Br J Haematol 2017; 176(4): 600–608
https://doi.org/10.1111/bjh.14455
15 J Wu, Y Li, J Wu, H Song, Y Tang, N Yan, L Wu, S Zhang, C Chang, M Lu. Decitabine activates type I interferon signaling to inhibit p53-deficient myeloid malignant cells. Clin Transl Med 2021; 11(11): e593
https://doi.org/10.1002/ctm2.593
16 JS Welch, AA Petti, CA Miller, CC Fronick, M O’Laughlin, RS Fulton, RK Wilson, JD Baty, EJ Duncavage, B Tandon, YS Lee, LD Wartman, GL Uy, A Ghobadi, MH Tomasson, I Pusic, R Romee, TA Fehniger, KE Stockerl-Goldstein, R Vij, ST Oh, CN Abboud, AF Cashen, MA Schroeder, MA Jacoby, SE Heath, K Luber, MR Janke, A Hantel, N Khan, MJ Sukhanova, RW Knoebel, W Stock, TA Graubert, MJ Walter, P Westervelt, DC Link, JF DiPersio, TJ Ley. TP53 and decitabine in acute myeloid leukemia and myelodysplastic syndromes. N Engl J Med 2016; 375(21): 2023–2036
https://doi.org/10.1056/NEJMoa1605949
17 K Appleton, HJ Mackay, I Judson, JA Plumb, C McCormick, G Strathdee, C Lee, S Barrett, S Reade, D Jadayel, A Tang, K Bellenger, L Mackay, A Setanoians, A Schätzlein, C Twelves, SB Kaye, R Brown. Phase I and pharmacodynamic trial of the DNA methyltransferase inhibitor decitabine and carboplatin in solid tumors. J Clin Oncol 2007; 25(29): 4603–4609
https://doi.org/10.1200/JCO.2007.10.8688
18 WE Samlowski, SA Leachman, M Wade, P Cassidy, P Porter-Gill, L Busby, R Wheeler, K Boucher, F Fitzpatrick, DA Jones, AR Karpf. Evaluation of a 7-day continuous intravenous infusion of decitabine: inhibition of promoter-specific and global genomic DNA methylation. J Clin Oncol 2005; 23(17): 3897–3905
https://doi.org/10.1200/JCO.2005.06.118
19 X Fu, Y Zhang, X Wang, M Chen, Y Wang, J Nie, Y Meng, W Han. Low dose decitabine combined with taxol and platinum chemotherapy to treat refractory/recurrent ovarian cancer: an open-label, single-arm, phase I/II study. Curr Protein Pept Sci 2015; 16(4): 329–336
https://doi.org/10.2174/138920371604150429155740
20 D Matei, F Fang, C Shen, J Schilder, A Arnold, Y Zeng, WA Berry, T Huang, KP Nephew. Epigenetic resensitization to platinum in ovarian cancer. Cancer Res 2012; 72(9): 2197–2205
https://doi.org/10.1158/0008-5472.CAN-11-3909
21 Y Zhang, Q Mei, Y Liu, X Li, MV Brock, M Chen, L Dong, L Shi, Y Wang, M Guo, J Nie, W Han. The safety, efficacy, and treatment outcomes of a combination of low-dose decitabine treatment in patients with recurrent ovarian cancer. Oncoimmunology 2017; 6(9): e1323619
https://doi.org/10.1080/2162402X.2017.1323619
22 A van der Westhuizen, N Knoblauch, MC Graves, R Levy, RE Vilain, NA Bowden. Pilot early phase II study of decitabine and carboplatin in patients with advanced melanoma. Medicine (Baltimore) 2020; 99(25): e20705
https://doi.org/10.1097/MD.0000000000020705
23 A Stathis, SJ Hotte, EX Chen, HW Hirte, AM Oza, P Moretto, S Webster, A Laughlin, LA Stayner, S McGill, L Wang, WJ Zhang, I Espinoza-Delgado, JL Holleran, MJ Egorin, LL Siu. Phase I study of decitabine in combination with vorinostat in patients with advanced solid tumors and non-Hodgkin’s lymphomas. Clin Cancer Res 2011; 17(6): 1582–1590
https://doi.org/10.1158/1078-0432.CCR-10-1893
24 B Leroy, M Anderson, T Soussi. TP53 mutations in human cancer: database reassessment and prospects for the next decade. Hum Mutat 2014; 35(6): 672–688
https://doi.org/10.1002/humu.22552
25 F Fang, C Balch, J Schilder, T Breen, S Zhang, C Shen, L Li, C Kulesavage, AJ Snyder, KP Nephew, DE Matei. A phase 1 and pharmacodynamic study of decitabine in combination with carboplatin in patients with recurrent, platinum-resistant, epithelial ovarian cancer. Cancer 2010; 116(17): 4043–4053
https://doi.org/10.1002/cncr.25204
26 J Yu, B Qin, AM Moyer, S Nowsheen, T Liu, S Qin, Y Zhuang, D Liu, SW Lu, KR Kalari, DW Visscher, JA Copland, SA McLaughlin, A Moreno-Aspitia, DW Northfelt, RJ Gray, Z Lou, VJ Suman, R Weinshilboum, JC Boughey, MP Goetz, L Wang. DNA methyltransferase expression in triple-negative breast cancer predicts sensitivity to decitabine. J Clin Invest 2018; 128(6): 2376–2388
https://doi.org/10.1172/JCI97924
27 A Stathis, SJ Hotte, EX Chen, HW Hirte, AM Oza, P Moretto, S Webster, A Laughlin, LA Stayner, S McGill, L Wang, WJ Zhang, I Espinoza-Delgado, JL Holleran, MJ Egorin, LL Siu. Phase I study of decitabine in combination with vorinostat in patients with advanced solid tumors and non-Hodgkin’s lymphomas. Clin Cancer Res 2011; 17(6): 1582–1590
https://doi.org/10.1158/1078-0432.CCR-10-1893
28 K Patel, J Dickson, S Din, K Macleod, D Jodrell, B Ramsahoye. Targeting of 5-aza-2′-deoxycytidine residues by chromatin-associated DNMT1 induces proteasomal degradation of the free enzyme. Nucleic Acids Res 2010; 38(13): 4313–4324
https://doi.org/10.1093/nar/gkq187
29 F Creusot, G Acs, JK Christman. Inhibition of DNA methyltransferase and induction of Friend erythroleukemia cell differentiation by 5-azacytidine and 5-aza-2′-deoxycytidine. J Biol Chem 1982; 257(4): 2041–2048
https://doi.org/10.1016/S0021-9258(19)68144-5
30 DJ Stewart, JP Issa, R Kurzrock, MI Nunez, J Jelinek, D Hong, Y Oki, Z Guo, S Gupta, II Wistuba. Decitabine effect on tumor global DNA methylation and other parameters in a phase I trial in refractory solid tumors and lymphomas. Clin Cancer Res 2009; 15(11): 3881–3888
https://doi.org/10.1158/1078-0432.CCR-08-2196
31 J Yu, B Qin, AM Moyer, S Nowsheen, T Liu, S Qin, Y Zhuang, D Liu, SW Lu, KR Kalari, DW Visscher, JA Copland, SA McLaughlin, A Moreno-Aspitia, DW Northfelt, RJ Gray, Z Lou, VJ Suman, R Weinshilboum, JC Boughey, MP Goetz, L Wang. DNA methyltransferase expression in triple-negative breast cancer predicts sensitivity to decitabine. J Clin Invest 2018; 128(6): 2376–2388
https://doi.org/10.1172/JCI97924
32 M Köbel, A Reuss, Bois A du, S Kommoss, F Kommoss, D Gao, SE Kalloger, DG Huntsman, CB Gilks. The biological and clinical value of p53 expression in pelvic high-grade serous carcinomas. J Pathol 2010; 222(2): 191–198
https://doi.org/10.1002/path.2744
33 A Talhouk, MK McConechy, S Leung, W Yang, A Lum, J Senz, N Boyd, J Pike, M Anglesio, JS Kwon, AN Karnezis, DG Huntsman, CB Gilks, JN McAlpine. Confirmation of ProMisE: a simple, genomics-based clinical classifier for endometrial cancer. Cancer 2017; 123(5): 802–813
https://doi.org/10.1002/cncr.30496
34 A Talhouk, MK McConechy, S Leung, HH Li-Chang, JS Kwon, N Melnyk, W Yang, J Senz, N Boyd, AN Karnezis, DG Huntsman, CB Gilks, JN McAlpine. A clinically applicable molecular-based classification for endometrial cancers. Br J Cancer 2015; 113(2): 299–310
https://doi.org/10.1038/bjc.2015.190
35 Genome Atlas Network Cancer. Comprehensive molecular portraits of human breast tumours. Nature 2012; 490(7418): 61–70
https://doi.org/10.1038/nature11412
36 B Pereira, SF Chin, OM Rueda, HKM Vollan, E Provenzano, HA Bardwell, M Pugh, L Jones, R Russell, SJ Sammut, DWY Tsui, B Liu, SJ Dawson, J Abraham, H Northen, JF Peden, A Mukherjee, G Turashvili, AR Green, S McKinney, A Oloumi, S Shah, N Rosenfeld, L Murphy, DR Bentley, IO Ellis, A Purushotham, SE Pinder, AL Børresen-Dale, HM Earl, PD Pharoah, MT Ross, S Aparicio, C Caldas. Erratum: The somatic mutation profiles of 2433 breast cancers refine their genomic and transcriptomic landscapes. Nat Commun 2016; 7(1): 11908
https://doi.org/10.1038/ncomms11479
37 R Maisano, M Zavettieri, D Azzarello, M Raffaele, M Maisano, M Bottari, M Nardi. Carboplatin and gemcitabine combination in metastatic triple-negative anthracycline- and taxane-pretreated breast cancer patients: a phase II study. J Chemother 2011; 23(1): 40–43
https://doi.org/10.1179/joc.2011.23.1.40
38 A Tutt, H Tovey, MCU Cheang, S Kernaghan, L Kilburn, P Gazinska, J Owen, J Abraham, S Barrett, P Barrett-Lee, R Brown, S Chan, M Dowsett, JM Flanagan, L Fox, A Grigoriadis, A Gutin, C Harper-Wynne, MQ Hatton, KA Hoadley, J Parikh, P Parker, CM Perou, R Roylance, V Shah, A Shaw, IE Smith, KM Timms, AM Wardley, G Wilson, C Gillett, JS Lanchbury, A Ashworth, N Rahman, M Harries, P Ellis, SE Pinder, JM Bliss. Carboplatin in BRCA1/2-mutated and triple-negative breast cancer BRCAness subgroups: the TNT Trial. Nat Med 2018; 24(5): 628–637
https://doi.org/10.1038/s41591-018-0009-7
39 J O’Shaughnessy, L Schwartzberg, MA Danso, KD Miller, HS Rugo, M Neubauer, N Robert, B Hellerstedt, M Saleh, P Richards, JM Specht, DA Yardley, RW Carlson, RS Finn, E Charpentier, I Garcia-Ribas, EP Winer. Phase III study of iniparib plus gemcitabine and carboplatin versus gemcitabine and carboplatin in patients with metastatic triple-negative breast cancer. J Clin Oncol 2014; 32(34): 3840–3847
https://doi.org/10.1200/JCO.2014.55.2984
40 SJ Isakoff, EL Mayer, L He, TA Traina, LA Carey, KJ Krag, HS Rugo, MC Liu, V Stearns, SE Come, KM Timms, AR Hartman, DR Borger, DM Finkelstein, JE Garber, PD Ryan, EP Winer, PE Goss, LW Ellisen. TBCRC009: a multicenter phase II clinical trial of platinum monotherapy with biomarker assessment in metastatic triple-negative breast cancer. J Clin Oncol 2015; 33(17): 1902–1909
https://doi.org/10.1200/JCO.2014.57.6660
41 LA Carey, HS Rugo, PK Marcom, EL Mayer, FJ Esteva, CX Ma, MC Liu, AM Storniolo, MF Rimawi, A Forero-Torres, AC Wolff, TJ Hobday, A Ivanova, WK Chiu, M Ferraro, E Burrows, PS Bernard, KA Hoadley, CM Perou, EP Winer. TBCRC 001: randomized phase II study of cetuximab in combination with carboplatin in stage IV triple-negative breast cancer. J Clin Oncol 2012; 30(21): 2615–2623
https://doi.org/10.1200/JCO.2010.34.5579
42 J Wu, H Song, Z Wang, M Lu. Three optimized assays for the evaluation of compounds that can rescue p53 mutants. STAR Protoc 2021; 2(3): 100688
https://doi.org/10.1016/j.xpro.2021.100688
43 JK Christman. 5-Azacytidine and 5-aza-2′-deoxycytidine as inhibitors of DNA methylation: mechanistic studies and their implications for cancer therapy. Oncogene 2002; 21(35): 5483–5495
https://doi.org/10.1038/sj.onc.1205699
44 C Stresemann, F Lyko. Modes of action of the DNA methyltransferase inhibitors azacytidine and decitabine. Int J Cancer 2008; 123(1): 8–13
https://doi.org/10.1002/ijc.23607
45 CA Brady, D Jiang, SS Mello, TM Johnson, LA Jarvis, MM Kozak, D Kenzelmann Broz, S Basak, EJ Park, ME McLaughlin, AN Karnezis, LD Attardi. Distinct p53 transcriptional programs dictate acute DNA-damage responses and tumor suppression. Cell 2011; 145(4): 571–583
https://doi.org/10.1016/j.cell.2011.03.035
46 M Olivier, M Hollstein, P Hainaut. TP53 mutations in human cancers: origins, consequences, and clinical use. Cold Spring Harb Perspect Biol 2010; 2(1): a001008
https://doi.org/10.1101/cshperspect.a001008
47 S Bhattacharya, P Dunn, CG Thomas, B Smith, H Schaefer, J Chen, Z Hu, KA Zalocusky, RD Shankar, SS Shen-Orr, E Thomson, J Wiser, AJ Butte. ImmPort, toward repurposing of open access immunological assay data for translational and clinical research. Sci Data 2018; 5(1): 180015
https://doi.org/10.1038/sdata.2018.15
48 R Colina, M Costa-Mattioli, RJ Dowling, M Jaramillo, LH Tai, CJ Breitbach, Y Martineau, O Larsson, L Rong, YV Svitkin, AP Makrigiannis, JC Bell, N Sonenberg. Translational control of the innate immune response through IRF-7. Nature 2008; 452(7185): 323–328
https://doi.org/10.1038/nature06730
49 M Sgarbanti, G Marsili, AL Remoli, R Orsatti, A Battistini. IRF-7: new role in the regulation of genes involved in adaptive immunity. Ann N Y Acad Sci 2007; 1095(1): 325–333
https://doi.org/10.1196/annals.1397.036
50 R Lu, WC Au, WS Yeow, N Hageman, PM Pitha. Regulation of the promoter activity of interferon regulatory factor-7 gene. Activation by interferon snd silencing by hypermethylation. J Biol Chem 2000; 275(41): 31805–31812
https://doi.org/10.1074/jbc.M005288200
51 J Yu, HY Zhang, ZZ Ma, W Lu, YF Wang, JD Zhu. Methylation profiling of twenty four genes and the concordant methylation behaviours of nineteen genes that may contribute to hepatocellular carcinogenesis. Cell Res 2003; 13(5): 319–333
https://doi.org/10.1038/sj.cr.7290177
52 Q Li, L Tang, PC Roberts, JM Kraniak, AL Fridman, OI Kulaeva, OS Tehrani, MA Tainsky. Interferon regulatory factors IRF5 and IRF7 inhibit growth and induce senescence in immortal Li-Fraumeni fibroblasts. Mol Cancer Res 2008; 6(5): 770–784
https://doi.org/10.1158/1541-7786.MCR-07-0114
[1] Hongnan Mo, Binghe Xu. Progress in systemic therapy for triple-negative breast cancer[J]. Front. Med., 2021, 15(1): 1-10.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed