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 (4) : 721-734    https://doi.org/10.1007/s11684-023-1053-3
Epigenetic silencing of BEND4, a novel DNA damage repair gene, is a synthetic lethal marker for ATM inhibitor in pancreatic cancer
Yuanxin Yao1, Honghui Lv2,1, Meiying Zhang1, Yuan Li3,1, James G. Herman4, Malcolm V. Brock5, Aiai Gao1, Qian Wang1, Francois Fuks6, Lirong Zhang2(), Mingzhou Guo1,7()
1. Department of Gastroenterology and Hepatology, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
2. Department of Pharmacology, School of Basic Medical Sciences, Zhengzhou University, Zhengzhou 450001, China
3. Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou 450001, China
4. UPMC Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA 15213, USA
5. Department of surgery, School of Medicine, Johns Hopkins School of Medicine, Baltimore, MD 21287, USA
6. Laboratory of Cancer Epigenetics, Faculty of Medicine, ULB-Cancer Research Center (U-CRC), Universite Libre de Bruxelles (ULB), Brussels 1070, Belgium
7. National Key Laboratory of Kidney Diseases, the First Medical Center, Chinese PLA General Hospital, Beijing 100853, China
 Download: PDF(16163 KB)   HTML
 Export: BibTeX | EndNote | Reference Manager | ProCite | RefWorks
Abstract

Synthetic lethality is a novel model for cancer therapy. To understand the function and mechanism of BEN domain-containing protein 4 (BEND4) in pancreatic cancer, eight cell lines and a total of 492 cases of pancreatic neoplasia samples were included in this study. Methylation-specific polymerase chain reaction, CRISPR/Cas9, immunoprecipitation assay, comet assay, and xenograft mouse model were used. BEND4 is a new member of the BEN domain family. The expression of BEND4 is regulated by promoter region methylation. It is methylated in 58.1% (176/303) of pancreatic ductal adenocarcinoma (PDAC), 33.3% (14/42) of intraductal papillary mucinous neoplasm, 31.0% (13/42) of pancreatic neuroendocrine tumor, 14.3% (3/21) of mucinous cystic neoplasm, 4.3% (2/47) of solid pseudopapillary neoplasm, and 2.7% (1/37) of serous cystic neoplasm. BEND4 methylation is significantly associated with late-onset PDAC (> 50 years, P < 0.01) and tumor differentiation (P < 0.0001), and methylation of BEND4 is an independent poor prognostic marker (P < 0.01) in PDAC. Furthermore, BEND4 plays tumor-suppressive roles in vitro and in vivo. Mechanistically, BEND4 involves non-homologous end joining signaling by interacting with Ku80 and promotes DNA damage repair. Loss of BEND4 increased the sensitivity of PDAC cells to ATM inhibitor. Collectively, the present study revealed an uncharacterized tumor suppressor BEND4 and indicated that methylation of BEND4 may serve as a potential synthetic lethal marker for ATM inhibitor in PDAC treatment.

Keywords BEND4      DNA methylation      synthetic lethality      NHEJ pathway     
Corresponding Author(s): Lirong Zhang,Mingzhou Guo   
Just Accepted Date: 16 May 2024   Online First Date: 28 June 2024    Issue Date: 30 August 2024
 Cite this article:   
Yuanxin Yao,Honghui Lv,Meiying Zhang, et al. Epigenetic silencing of BEND4, a novel DNA damage repair gene, is a synthetic lethal marker for ATM inhibitor in pancreatic cancer[J]. Front. Med., 2024, 18(4): 721-734.
 URL:  
https://academic.hep.com.cn/fmd/EN/10.1007/s11684-023-1053-3
https://academic.hep.com.cn/fmd/EN/Y2024/V18/I4/721
Fig.1  Expression and methylation status of BEND4 in PDAC cells and tissue samples. (A) The association of BEND4 expression and CpG site methylation in the promoter region extracted from the TCGA database. (B) RT-PCR and (C) MSP results of BEND4 in PDAC cells. 5-aza: 5-aza-2′-deoxycytidine; GAPDH: internal control; U: unmethylation; M: methylation; IVD: in vitro methylated DNA, serves as methylation control; NL: normal peripheral lymphocytes DNA, serves as unmethylation control. (D) BSSQ results of BEND4 in PDAC cells. MSP product size is 143 bp, and BSSQ was amplified for 301 bp (from −324 to −23) encompassing the MSP product in the promoter region. Filled circles: methylated CpG sites; open circles: unmethylated CpG sites; TSS: transcription start site. (E) Representative MSP results of BEND4 in pancreatic neoplasms. PN: noncancerous pancreatic tissue samples; IPMN: intraductal papillary mucinous neoplasm; PDAC: pancreatic ductal adenocarcinoma. (F) Representative IHC staining of BEND4 in PDAC and adjacent noncancerous tissue samples. Scale bar: 50 μm (up); 20 μm (down). (G) IHC score of BEND4 in PDAC and adjacent noncancerous tissue samples. Data are presented as mean ± SD. (H) The association of BEND4 expression and promoter region methylation in PDAC. (I) Kaplan–Meier curve shows the association of BEND4 methylation and OS in patients with PDAC (n = 207). ****P < 0.0001.
Variables Number BEND4 methylation status P
Unmethylation n = 127 (41.9%) Methylation n = 176 (58.1%)
Age (year) 0.0083**
≤ 50 51 30 21
> 50 252 97 155
Gender 0.904
Female 111 46 65
Male 192 81 111
Smoking 0.055
No 185 86 99
Yes 118 41 77
Alcohol abuse > 0.999
No 165 69 96
Yes 138 58 80
Tumor size (cm) 0.483
≤ 4 237 102 135
> 4 66 25 41
Tumor location > 0.999
Head and neck 143 60 83
Body and tail 160 67 93
Differentiation < 0.0001****
Well or moderate 137 75 62
Poor 157 50 107
TNM stage 0.842
Stage I–II 275 116 159
Stage III–IV 28 11 17
Lymph node metastasis 0.067
No 199 91 108
Yes 104 36 68
Nerve invasion > 0.999
No 81 34 47
Yes 222 93 129
Tab.1  Association of BEND4 methylation and clinical factors in PDAC
Variables Univariate analysis Multivariate analysis
HR (95% CI) P HR (95% CI) P
Gender
Male vs female 0.858 (0.545, 1.351) 0.509
Age (year)
> 50 vs ≤ 50 1.506 (0.775, 2.927) 0.227
Size (cm)
> 4 vs ≤ 4 1.411 (0.849, 2.347) 0.184
Differentiation
Well or moderate vs poor 0.642 (0.407, 1.013) 0.057
Lymph node metastasis
Positive vs negative 1.748 (1.124, 2.718) 0.013* 1.598 (1.025, 2.490) 0.038*
TNM stage
III + IV vs I + II 1.270 (0.687, 2.349) 0.445
BEND4
Methylation vs unmethylation 2.315 (1.443, 3.714) 0.001** 2.196 (1.365, 3.532) 0.001**
Tab.2  Univariate and multivariate analyses of BEND4 methylation status with the overall survival of patients with PDAC
Fig.2  Effect of BEND4 on PDAC cell proliferation, colony formation, cell cycle, apoptosis, migration, invasion, and tumor xenograft model. (A) Cell proliferation was evaluated by MTT assay. Vector: empty vector control; BEND4: BEND4 expressing vector. WT: wild-type control; KO: BEND4 knockout. (B) Representative colony formation results. (C) Flow cytometry shows cell cycle results. (D) Levels of checkpoint proteins in PDAC cells without or with BEND4 expression. β-actin: internal control. (E) BEND4 induces apoptosis in PDAC cells. Scr: scramble control. (F) Levels of key apoptotic proteins in PDAC cells without or with BEND4 expression. (G) BEND4 suppresses migration and invasion in PDAC cells. (H) Levels of metastatic proteins in PDAC cells without or with BEND4 expression. (I) Representative pictures of BEND4 unexpressed and re-expressed SW1990 cell xenografts. (J) Growth curves and (K) Tumor weight of BEND4 unexpressed and re-expressed SW1990 cell xenografts (n = 6). The results are presented as the mean ± SD of three replicates. *P < 0.05, ***P < 0.001, ****P < 0.0001.
Fig.3  BEND4 promotes DNA damage repair through NHEJ. (A) Co-IP assay and silver staining. Red arrow: differential band. (B) Co-IP (up) and reciprocal Co-IP (down) validation of BEND4 binding with Ku80. (C) Comet assay in PDAC cells before and after cisplatin treatment. (D) Levels of NHEJ proteins in PDAC cells before and after cisplatin treatment. The results are presented as the mean ± SD of three replicates. ns, no significance. ****P < 0.0001.
Fig.4  BEND4 involves the NHEJ signaling pathway by interacting with Ku80. (A) HR and NHEJ efficiency were evaluated in U2OS-DR-GFP and U2OS-EJ5 cells before and after BEND4 knockdown. (B) Co-IP assay to show the interaction of Ku70, Ku80, and DNAPKcs in BEND4 unexpressed and overexpressed SW1990 cells. (C) Ku70, Ku80, and DNAPKcs in whole-cell lysates and chromatin fraction of PDAC cells after cisplatin treatment. (D) Comet assay in PDAC cells before and after knockdown of Ku80. (E) Levels of NHEJ proteins before and after Ku80 knockdown. The results are presented as the mean ± SD of three replicates. ns, no significance. *P < 0.05, ****P < 0.0001.
Fig.5  Loss of BEND4 expression sensitized PDAC cells to AZD0156. (A) IC50 assay to show the sensitivity of PDAC cells to AZD0156. (B) Representative colony formation results to show the synthetic lethality between AZD0156 and BEND4 defect. (C) Levels of ATM/CHK2 signaling and γ-H2AX in cells under different combination of treatment with cisplatin and AZD0156. The results are presented as the mean ± SD of three replicates. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.
Fig.6  Synthetic lethality of BEND4 methylation and ATM inhibitor in vivo. (A) Tumor growth curves of BEND4 unexpressed and re-expressed SW1990 cell xenografts under different combinations of treatment with cisplatin and AZD0156. (B) Normalized tumor volume and (C) weight in BEND4 unexpressed and re-expressed SW1990 cell xenografts. (D) Representative IHC results to show the levels of NHEJ proteins in SW1990 cell xenografts under cisplatin treatment. (E) A working model for synthetic lethality of BEND4 methylation and ATM inhibitor in PDAC cells. M: DNA methylation; NHEJ: non-homologous end joining; HR: homologous recombination. ***P < 0.001, ****P < 0.0001.
1 L Rahib, BD Smith, R Aizenberg, AB Rosenzweig, JM Fleshman, LM Matrisian. Projecting cancer incidence and deaths to 2030: the unexpected burden of thyroid, liver, and pancreas cancers in the United States. Cancer Res 2014; 74(11): 2913–2921
https://doi.org/10.1158/0008-5472.CAN-14-0155
2 JD Mizrahi, R Surana, JW Valle, RT Shroff. Pancreatic cancer. Lancet 2020; 395(10242): 2008–2020
https://doi.org/10.1016/S0140-6736(20)30974-0
3 O Strobel, J Neoptolemos, D Jäger, MW Büchler. Optimizing the outcomes of pancreatic cancer surgery. Nat Rev Clin Oncol 2019; 16(1): 11–26
https://doi.org/10.1038/s41571-018-0112-1
4 SB Dreyer, DK Chang, P Bailey, AV Biankin. Pancreatic cancer genomes: implications for clinical management and therapeutic development. Clin Cancer Res 2017; 23(7): 1638–1646
https://doi.org/10.1158/1078-0432.CCR-16-2411
5 A Huang, LA Garraway, A Ashworth, B Weber. Synthetic lethality as an engine for cancer drug target discovery. Nat Rev Drug Discov 2020; 19(1): 23–38
https://doi.org/10.1038/s41573-019-0046-z
6 H Farmer, N McCabe, CJ Lord, AN Tutt, DA Johnson, TB Richardson, M Santarosa, KJ Dillon, I Hickson, C Knights, NM Martin, SP Jackson, GC Smith, A Ashworth. Targeting the DNA repair defect in BRCA mutant cells as a therapeutic strategy. Nature 2005; 434(7035): 917–921
https://doi.org/10.1038/nature03445
7 HE Bryant, N Schultz, HD Thomas, KM Parker, D Flower, E Lopez, S Kyle, M Meuth, NJ Curtin, T Helleday. Specific killing of BRCA2-deficient tumours with inhibitors of poly(ADP-ribose) polymerase. Nature 2005; 434(7035): 913–917
https://doi.org/10.1038/nature03443
8 CJ Lord, A Ashworth. PARP inhibitors: synthetic lethality in the clinic. Science 2017; 355(6330): 1152–1158
https://doi.org/10.1126/science.aam7344
9 CJ Lord, A Ashworth. BRCAness revisited. Nat Rev Cancer 2016; 16(2): 110–120
https://doi.org/10.1038/nrc.2015.21
10 M Guo, Y Peng, A Gao, C Du, JG Herman. Epigenetic heterogeneity in cancer. Biomark Res 2019; 7(1): 23
https://doi.org/10.1186/s40364-019-0174-y
11 A Gao, M Guo. Epigenetic based synthetic lethal strategies in human cancers. Biomark Res 2020; 8: 44
https://doi.org/10.1186/s40364-020-00224-1
12 Y Cui, H Chen, R Xi, H Cui, Y Zhao, E Xu, T Yan, X Lu, F Huang, P Kong, Y Li, X Zhu, J Wang, W Zhu, J Wang, Y Ma, Y Zhou, S Guo, L Zhang, Y Liu, B Wang, Y Xi, R Sun, X Yu, Y Zhai, F Wang, J Yang, B Yang, C Cheng, J Liu, B Song, H Li, Y Wang, Y Zhang, X Cheng, Q Zhan, Y Li, Z Liu. Whole-genome sequencing of 508 patients identifies key molecular features associated with poor prognosis in esophageal squamous cell carcinoma. Cell Res 2020; 30(10): 902–913
https://doi.org/10.1038/s41422-020-0333-6
13 S Abhiman, LM Iyer, L Aravind. BEN: a novel domain in chromatin factors and DNA viral proteins. Bioinformatics 2008; 24(4): 458–461
https://doi.org/10.1093/bioinformatics/btn007
14 KM Sathyan, Z Shen, V Tripathi, KV Prasanth, SG Prasanth. A BEN-domain-containing protein associates with heterochromatin and represses transcription. J Cell Sci 2011; 124(Pt 18): 3149–3163
https://doi.org/10.1242/jcs.086603
15 F Kurniawan, SG Prasanth. A BEN-domain protein and polycomb complex work coordinately to regulate transcription. Transcription 2022; 13(1–3): 82–87
https://doi.org/10.1080/21541264.2022.2105128
16 J Zhang, Y Zhang, Q You, C Huang, T Zhang, M Wang, T Zhang, X Yang, J Xiong, Y Li, CP Liu, Z Zhang, RM Xu, B Zhu. Highly enriched BEND3 prevents the premature activation of bivalent genes during differentiation. Science 2022; 375(6584): 1053–1058
https://doi.org/10.1126/science.abm0730
17 S Babu, Y Takeuchi, I Masai. Banp regulates DNA damage response and chromosome segregation during the cell cycle in zebrafish retina. eLife 2022; 11: e74611
https://doi.org/10.7554/eLife.74611
18 L Ma, D Xie, M Luo, X Lin, H Nie, J Chen, C Gao, S Duo, C Han. Identification and characterization of BEND2 as a key regulator of meiosis during mouse spermatogenesis. Sci Adv 2022; 8(21): eabn1606
https://doi.org/10.1126/sciadv.abn1606
19 Y Shi, D Zhang, J Chen, Q Jiang, S Song, Y Mi, T Wang, Q Ye. Interaction between BEND5 and RBPJ suppresses breast cancer growth and metastasis via inhibiting Notch signaling. Int J Biol Sci 2022; 18(10): 4233–4244
https://doi.org/10.7150/ijbs.70866
20 G Shi, Y Bai, X Zhang, J Su, J Pang, Q He, P Zeng, J Ding, Y Xiong, J Zhang, J Wang, D Liu, W Ma, J Huang, Z Songyang. Bend family proteins mark chromatin boundaries and synergistically promote early germ cell differentiation. Protein Cell 2022; 13(10): 721–741
https://doi.org/10.1007/s13238-021-00884-1
21 W Du, A Gao, JG Herman, L Wang, L Zhang, S Jiao, M Guo. Methylation of NRN1 is a novel synthetic lethal marker of PI3K-Akt-mTOR and ATR inhibitors in esophageal cancer. Cancer Sci 2021; 112(7): 2870–2883
https://doi.org/10.1111/cas.14917
22 H Li, M Zhang, E Linghu, F Zhou, JG Herman, L Hu, M Guo. Epigenetic silencing of TMEM176A activates ERK signaling in human hepatocellular carcinoma. Clin Epigenetics 2018; 10(1): 137
https://doi.org/10.1186/s13148-018-0570-4
23 AJ Pierce, RD Johnson, LH Thompson, M Jasin. XRCC3 promotes homology-directed repair of DNA damage in mammalian cells. Genes Dev 1999; 13(20): 2633–2638
https://doi.org/10.1101/gad.13.20.2633
24 J Guirouilh-Barbat, S Huck, P Bertrand, L Pirzio, C Desmaze, L Sabatier, BS Lopez. Impact of the KU80 pathway on NHEJ-induced genome rearrangements in mammalian cells. Mol Cell 2004; 14(5): 611–623
https://doi.org/10.1016/j.molcel.2004.05.008
25 PL Olive, JP Banáth. The comet assay: a method to measure DNA damage in individual cells. Nat Protoc 2006; 1(1): 23–29
https://doi.org/10.1038/nprot.2006.5
26 C Zhang, B Zhou, F Gu, H Liu, H Wu, F Yao, H Zheng, H Fu, W Chong, S Cai, M Huang, X Ma, Z Guo, T Li, W Deng, M Zheng, Q Ji, Y Zhao, Y Ma, QE Wang, TS Tang, C Guo. Micropeptide PACMP inhibition elicits synthetic lethal effects by decreasing CtIP and poly(ADP-ribosyl)ation. Mol Cell 2022; 82(7): 1297–1312.e8
https://doi.org/10.1016/j.molcel.2022.01.020
27 B Zhao, E Rothenberg, DA Ramsden, MR Lieber. The molecular basis and disease relevance of non-homologous DNA end joining. Nat Rev Mol Cell Biol 2020; 21(12): 765–781
https://doi.org/10.1038/s41580-020-00297-8
28 X Yue, C Bai, D Xie, T Ma, PK Zhou. DNA-PKcs: a multi-faceted player in DNA damage response. Front Genet 2020; 11: 607428
https://doi.org/10.3389/fgene.2020.607428
29 C Almoguera, D Shibata, K Forrester, J Martin, N Arnheim, M Perucho. Most human carcinomas of the exocrine pancreas contain mutant c-K-ras genes. Cell 1988; 53(4): 549–554
https://doi.org/10.1016/0092-8674(88)90571-5
30 A Scarpa, P Capelli, K Mukai, G Zamboni, T Oda, C Iacono, S Hirohashi. Pancreatic adenocarcinomas frequently show p53 gene mutations. Am J Pathol 1993; 142(5): 1534–1543
31 S Jones, X Zhang, DW Parsons, JC Lin, RJ Leary, P Angenendt, P Mankoo, H Carter, H Kamiyama, A Jimeno, SM Hong, B Fu, MT Lin, ES Calhoun, M Kamiyama, K Walter, T Nikolskaya, Y Nikolsky, J Hartigan, DR Smith, M Hidalgo, SD Leach, AP Klein, EM Jaffee, M Goggins, A Maitra, C Iacobuzio-Donahue, JR Eshleman, SE Kern, RH Hruban, R Karchin, N Papadopoulos, G Parmigiani, B Vogelstein, VE Velculescu, KW Kinzler. Core signaling pathways in human pancreatic cancers revealed by global genomic analyses. Science 2008; 321(5897): 1801–1806
https://doi.org/10.1126/science.1164368
32 AV Biankin, N Waddell, KS Kassahn, MC Gingras, LB Muthuswamy, AL Johns, DK Miller, PJ Wilson, AM Patch, J Wu, DK Chang, MJ Cowley, BB Gardiner, S Song, I Harliwong, S Idrisoglu, C Nourse, E Nourbakhsh, S Manning, S Wani, M Gongora, M Pajic, CJ Scarlett, AJ Gill, AV Pinho, I Rooman, M Anderson, O Holmes, C Leonard, D Taylor, S Wood, Q Xu, K Nones, JL Fink, A Christ, T Bruxner, N Cloonan, G Kolle, F Newell, M Pinese, RS Mead, JL Humphris, W Kaplan, MD Jones, EK Colvin, AM Nagrial, ES Humphrey, A Chou, VT Chin, LA Chantrill, A Mawson, JS Samra, JG Kench, JA Lovell, RJ Daly, ND Merrett, C Toon, K Epari, NQ Nguyen, A Barbour, N; Australian Pancreatic Cancer Genome Initiative; Kakkar N Zeps, F Zhao, YQ Wu, M Wang, DM Muzny, WE Fisher, FC Brunicardi, SE Hodges, JG Reid, J Drummond, K Chang, Y Han, LR Lewis, H Dinh, CJ Buhay, T Beck, L Timms, M Sam, K Begley, A Brown, D Pai, A Panchal, N Buchner, Borja R De, RE Denroche, CK Yung, S Serra, N Onetto, D Mukhopadhyay, MS Tsao, PA Shaw, GM Petersen, S Gallinger, RH Hruban, A Maitra, CA Iacobuzio-Donahue, RD Schulick, CL Wolfgang, RA Morgan, RT Lawlor, P Capelli, V Corbo, M Scardoni, G Tortora, MA Tempero, KM Mann, NA Jenkins, PA Perez-Mancera, DJ Adams, DA Largaespada, LF Wessels, AG Rust, LD Stein, DA Tuveson, NG Copeland, EA Musgrove, A Scarpa, JR Eshleman, TJ Hudson, RL Sutherland, DA Wheeler, JV Pearson, JD McPherson, RA Gibbs, SM Grimmond. Pancreatic cancer genomes reveal aberrations in axon guidance pathway genes. Nature 2012; 491(7424): 399–405
https://doi.org/10.1038/nature11547
33 P Bailey, DK Chang, K Nones, AL Johns, AM Patch, MC Gingras, DK Miller, AN Christ, TJ Bruxner, MC Quinn, C Nourse, LC Murtaugh, I Harliwong, S Idrisoglu, S Manning, E Nourbakhsh, S Wani, L Fink, O Holmes, V Chin, MJ Anderson, S Kazakoff, C Leonard, F Newell, N Waddell, S Wood, Q Xu, PJ Wilson, N Cloonan, KS Kassahn, D Taylor, K Quek, A Robertson, L Pantano, L Mincarelli, LN Sanchez, L Evers, J Wu, M Pinese, MJ Cowley, MD Jones, EK Colvin, AM Nagrial, ES Humphrey, LA Chantrill, A Mawson, J Humphris, A Chou, M Pajic, CJ Scarlett, AV Pinho, M Giry-Laterriere, I Rooman, JS Samra, JG Kench, JA Lovell, ND Merrett, CW Toon, K Epari, NQ Nguyen, A Barbour, N Zeps, K Moran-Jones, NB Jamieson, JS Graham, F Duthie, K Oien, J Hair, R Grützmann, A Maitra, CA Iacobuzio-Donahue, CL Wolfgang, RA Morgan, RT Lawlor, V Corbo, C Bassi, B Rusev, P Capelli, R Salvia, G Tortora, D Mukhopadhyay, GM; Australian Pancreatic Cancer Genome Initiative; Munzy DM Petersen, WE Fisher, SA Karim, JR Eshleman, RH Hruban, C Pilarsky, JP Morton, OJ Sansom, A Scarpa, EA Musgrove, UM Bailey, O Hofmann, RL Sutherland, DA Wheeler, AJ Gill, RA Gibbs, JV Pearson, N Waddell, AV Biankin, SM Grimmond. Genomic analyses identify molecular subtypes of pancreatic cancer. Nature 2016; 531(7592): 47–52
https://doi.org/10.1038/nature16965
34 SB Dreyer, R Upstill-Goddard, A Legrini, AV; Glasgow Precision Oncology Laboratory; Jamieson NB Biankin, DK; Australian Pancreatic Genome Initiative; Jamieson NB Chang, DK Chang. Genomic and molecular analyses identify molecular subtypes of pancreatic cancer recurrence. Gastroenterology 2022; 162(1): 320–324.e4
https://doi.org/10.1053/j.gastro.2021.09.022
35 N Waddell, M Pajic, AM Patch, DK Chang, KS Kassahn, P Bailey, AL Johns, D Miller, K Nones, K Quek, MC Quinn, AJ Robertson, MZ Fadlullah, TJ Bruxner, AN Christ, I Harliwong, S Idrisoglu, S Manning, C Nourse, E Nourbakhsh, S Wani, PJ Wilson, E Markham, N Cloonan, MJ Anderson, JL Fink, O Holmes, SH Kazakoff, C Leonard, F Newell, B Poudel, S Song, D Taylor, N Waddell, S Wood, Q Xu, J Wu, M Pinese, MJ Cowley, HC Lee, MD Jones, AM Nagrial, J Humphris, LA Chantrill, V Chin, AM Steinmann, A Mawson, ES Humphrey, EK Colvin, A Chou, CJ Scarlett, AV Pinho, M Giry-Laterriere, I Rooman, JS Samra, JG Kench, JA Pettitt, ND Merrett, C Toon, K Epari, NQ Nguyen, A Barbour, N Zeps, NB Jamieson, JS Graham, SP Niclou, R Bjerkvig, R Grützmann, D Aust, RH Hruban, A Maitra, CA Iacobuzio-Donahue, CL Wolfgang, RA Morgan, RT Lawlor, V Corbo, C Bassi, M Falconi, G Zamboni, G Tortora, MA; Australian Pancreatic Cancer Genome Initiative; Gill AJ Tempero, JR Eshleman, C Pilarsky, A Scarpa, EA Musgrove, JV Pearson, AV Biankin, SM Grimmond. Whole genomes redefine the mutational landscape of pancreatic cancer. Nature 2015; 518(7540): 495–501
https://doi.org/10.1038/nature14169
36 Genome Atlas Research Network Cancer. Integrated genomic characterization of pancreatic ductal adenocarcinoma. Cancer Cell 2017; 32(2): 185–203.e13
https://doi.org/10.1016/j.ccell.2017.07.007
37 AD Singhi, B George, JR Greenbowe, J Chung, J Suh, A Maitra, SJ Klempner, A Hendifar, JM Milind, T Golan, RE Brand, AH Zureikat, S Roy, AB Schrock, VA Miller, JS Ross, SM Ali, N Bahary. Real-time targeted genome profile analysis of pancreatic ductal adenocarcinomas identifies genetic alterations that might be targeted with existing drugs or used as biomarkers. Gastroenterology 2019; 156(8): 2242–2253.e4
https://doi.org/10.1053/j.gastro.2019.02.037
38 Y Qian, Y Gong, Z Fan, G Luo, Q Huang, S Deng, H Cheng, K Jin, Q Ni, X Yu, C Liu. Molecular alterations and targeted therapy in pancreatic ductal adenocarcinoma. J Hematol Oncol 2020; 13(1): 130
https://doi.org/10.1186/s13045-020-00958-3
39 MS Rémond, A Pellat, C Brezault, M Dhooge, R Coriat. Are targeted therapies or immunotherapies effective in metastatic pancreatic adenocarcinoma?. ESMO Open 2022; 7(6): 100638
https://doi.org/10.1016/j.esmoop.2022.100638
40 T Golan, P Hammel, M Reni, Cutsem E Van, T Macarulla, MJ Hall, JO Park, D Hochhauser, D Arnold, DY Oh, A Reinacher-Schick, G Tortora, H Algül, EM O’Reilly, D McGuinness, KY Cui, K Schlienger, GY Locker, HL Kindler. Maintenance olaparib for germline BRCA-mutated metastatic pancreatic cancer. N Engl J Med 2019; 381(4): 317–327
https://doi.org/10.1056/NEJMoa1903387
41 L Perkhofer, J Gout, E Roger, de Almeida F Kude, Simões C Baptista, L Wiesmüller, T Seufferlein, A Kleger. DNA damage repair as a target in pancreatic cancer: state-of-the-art and future perspectives. Gut 2021; 70(3): 606–617
https://doi.org/10.1136/gutjnl-2019-319984
42 A Hayashi, J Hong, CA Iacobuzio-Donahue. The pancreatic cancer genome revisited. Nat Rev Gastroenterol Hepatol 2021; 18(7): 469–481
https://doi.org/10.1038/s41575-021-00463-z
43 C Gourley, J Balmaña, JA Ledermann, V Serra, R Dent, S Loibl, E Pujade-Lauraine, SJ Boulton. Moving from Poly (ADP-ribose) polymerase inhibition to targeting DNA repair and DNA damage response in cancer therapy. J Clin Oncol 2019; 37(25): 2257–2269
https://doi.org/10.1200/JCO.18.02050
44 H Li, W Yang, M Zhang, T He, F Zhou, J G Herman, L Hu, M Guo. Methylation of TMEM176A, a key ERK signaling regulator, is a novel synthetic lethality marker of ATM inhibitors in human lung cancer. Epigenomics 2021; 13(17): 1403–1419
https://doi.org/10.2217/epi-2021-0217
45 V Davalos, M Esteller. Cancer epigenetics in clinical practice. CA Cancer J Clin 2023; 73(4): 376–424
https://doi.org/10.3322/caac.21765
46 MJ Topper, M Vaz, KA Marrone, JR Brahmer, SB Baylin. The emerging role of epigenetic therapeutics in immuno-oncology. Nat Rev Clin Oncol 2020; 17(2): 75–90
https://doi.org/10.1038/s41571-019-0266-5
47 D Wang, W Wu, E Callen, R Pavani, N Zolnerowich, S Kodali, D Zong, N Wong, S Noriega, WJ Nathan, G Matos-Rodrigues, R Chari, MJ Kruhlak, F Livak, M Ward, K Caldecott, B Di Stefano, A Nussenzweig. Active DNA demethylation promotes cell fate specification and the DNA damage response. Science 2022; 378(6623): 983–989
https://doi.org/10.1126/science.add9838
48 AR Weber, C Krawczyk, AB Robertson, A Kuśnierczyk, CB Vågbø, D Schuermann, A Klungland, P Schär. Biochemical reconstitution of TET1-TDG-BER-dependent active DNA demethylation reveals a highly coordinated mechanism. Nat Commun 2016; 7(1): 10806
https://doi.org/10.1038/ncomms10806
[1] FMD-23072-OF-GMZ_suppl_1 Download
[1] Lingyu Gao, Qianjin Lu. The critical importance of epigenetics in autoimmune-related skin diseases[J]. Front. Med., 2023, 17(1): 43-57.
[2] Jingming Li, Wen Jin, Yun Tan, Beichen Wang, Xiaoling Wang, Ming Zhao, Kankan Wang. Distinct gene expression pattern of RUNX1 mutations coordinated by target repression and promoter hypermethylation in acute myeloid leukemia[J]. Front. Med., 2022, 16(4): 627-636.
[3] Chen Wang, Hui Fang, Jiawei Zhang, Ying Gu. Targeting “undruggable” c-Myc protein by synthetic lethality[J]. Front. Med., 2021, 15(4): 541-550.
[4] Fusheng Zhou, Changbing Shen, Yi-Hsiang Hsu, Jing Gao, Jinfa Dou, Randy Ko, Xiaodong Zheng, Liangdan Sun, Yong Cui, Xuejun Zhang. DNA methylation-based subclassification of psoriasis in the Chinese Han population[J]. Front. Med., 2018, 12(6): 717-725.
[5] Yi Cao. Environmental pollution and DNA methylation: carcinogenesis, clinical significance, and practical applications[J]. Front. Med., 2015, 9(3): 261-274.
[6] Zhifeng Wang, Fengli Wang, Tieshan Tang, Caixia Guo. The role of PARP1 in the DNA damage response and its application in tumor therapy[J]. Front Med, 2012, 6(2): 156-164.
[7] Xiao-Mei ZHANG, Ming-Zhou GUO, . The value of epigenetic markers in esophageal cancer[J]. Front. Med., 2010, 4(4): 378-384.
[8] Hong LI, Shaoqin CHEN, Yi SHU, Yongjun CHEN, Ying SU, Xin WANG, Shengquan ZOU. Effects of hydralazine and valproate on the expression of E-cadherin gene and the invasiveness of QBC939 Cells[J]. Front Med Chin, 2009, 3(2): 153-157.
Viewed
Full text


Abstract

Cited

  Shared   
  Discussed