This work is licensed under the Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License.
Siegel, R. L., Miller, K. D., Fuchs, H. E. & Jemal, A. Cancer statistics, 2022. CA Cancer J Clin72, 7–33 (2022)SiegelR. L.MillerK. D.FuchsH. E.JemalA.Cancer statistics, 2022CA Cancer J Clin727332022Search in Google Scholar
Schouten, T. J. et al. Nationwide Validation of the 8th American Joint Committee on Cancer TNM Staging System and Five Proposed Modifications for Resected Pancreatic Cancer. Ann Surg Oncol29, 5988–5999 (2022)SchoutenT. J.Nationwide Validation of the 8th American Joint Committee on Cancer TNM Staging System and Five Proposed Modifications for Resected Pancreatic CancerAnn Surg Oncol29598859992022Search in Google Scholar
Balaban, E. P., Mangu, P. B. & Yee, N. S. Locally Advanced Unresectable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline Summary. J Oncol Pract13, 265–269 (2016)BalabanE. P.ManguP. B.YeeN. S.Locally Advanced Unresectable Pancreatic Cancer: American Society of Clinical Oncology Clinical Practice Guideline SummaryJ Oncol Pract132652692016Search in Google Scholar
Singh, R. R. & O’Reilly, E. M. New Treatment Strategies for Metastatic Pancreatic Ductal Adenocarcinoma. Drugs vol. 80 647–669 Preprint at https://doi.org/10.1007/s40265-020-01304-0 (2020)SinghR. R.O’ReillyE. M.New Treatment Strategies for Metastatic Pancreatic Ductal AdenocarcinomaDrugs80647669Preprint at https://doi.org/10.1007/s40265-020-01304-02020Search in Google Scholar
Elrakaybi, A., Ruess, D. A., Lübbert, M., Quante, M. & Becker, H. Epigenetics in Pancreatic Ductal Adenocarcinoma: Impact on Biology and Utilization in Diagnostics and Treatment. Cancers vol. 14 Preprint at https://doi.org/10.3390/cancers14235926 (2022)ElrakaybiA.RuessD. A.LübbertM.QuanteM.BeckerH.Epigenetics in Pancreatic Ductal Adenocarcinoma: Impact on Biology and Utilization in Diagnostics and TreatmentCancers14Preprint at https://doi.org/10.3390/cancers142359262022Search in Google Scholar
Sarantis, P., Koustas, E., Papadimitropoulou, A., Papavassiliou, A. G. & Karamouzis, M. V. Pancreatic ductal adenocarcinoma: Treatment hurdles, tumor microenvironment and immunotherapy. World Journal of Gastrointestinal Oncology vol. 12 173–181 Preprint at https://doi.org/10.4251/wjgo.v12.i2.173 (2020)SarantisP.KoustasE.PapadimitropoulouA.PapavassiliouA. G.KaramouzisM. V.Pancreatic ductal adenocarcinoma: Treatment hurdles, tumor microenvironment and immunotherapyWorld Journal of Gastrointestinal Oncology12173181Preprint at https://doi.org/10.4251/wjgo.v12.i2.1732020Search in Google Scholar
Siegel, R. L., Miller, K. D. & Jemal, A. Cancer statistics, 2020. CA Cancer J Clin70, 7–30 (2020)SiegelR. L.MillerK. D.JemalA.Cancer statistics, 2020CA Cancer J Clin707302020Search in Google Scholar
Von Hoff, D. D. et al. Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus Gemcitabine. New England Journal of Medicine369, 1691–1703 (2013)Von HoffD. D.Increased Survival in Pancreatic Cancer with nab-Paclitaxel plus GemcitabineNew England Journal of Medicine369169117032013Search in Google Scholar
Adamska, A. et al. Molecular and cellular mechanisms of chemoresistance in pancreatic cancer. Adv Biol Regul68, 77–87 (2018)AdamskaA.Molecular and cellular mechanisms of chemoresistance in pancreatic cancerAdv Biol Regul6877872018Search in Google Scholar
Sinn, M. et al. CONKO-005: Adjuvant Chemotherapy With Gemcitabine Plus Erlotinib Versus Gemcitabine Alone in Patients After R0 Resection of Pancreatic Cancer: A Multicenter Randomized Phase III Trial. Journal of Clinical Oncology35, 3330–3337 (2017)SinnM.CONKO-005: Adjuvant Chemotherapy With Gemcitabine Plus Erlotinib Versus Gemcitabine Alone in Patients After R0 Resection of Pancreatic Cancer: A Multicenter Randomized Phase III TrialJournal of Clinical Oncology35333033372017Search in Google Scholar
Conroy, T. et al. FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic Cancer. New England Journal of Medicine379, 2395–2406 (2018)ConroyT.FOLFIRINOX or Gemcitabine as Adjuvant Therapy for Pancreatic CancerNew England Journal of Medicine379239524062018Search in Google Scholar
Ma, W. W. & Hidalgo, M. The Winning Formulation: The Development of Paclitaxel in Pancreatic Cancer. Clinical Cancer Research19, 5572–5579 (2013)MaW. W.HidalgoM.The Winning Formulation: The Development of Paclitaxel in Pancreatic CancerClinical Cancer Research19557255792013Search in Google Scholar
Digiuseppe, J. A., Redston, M. S., Yeo, C. J., Kern, S. E. & Hruban, R. H. Communication P53-Independent Expression of the Cyclin-Dependent Kinase Inhibitor P21 in Pancreatic Carcinoma. American journal of Pathology vol. 147 (1995)DigiuseppeJ. A.RedstonM. S.YeoC. J.KernS. E.HrubanR. H.Communication P53-Independent Expression of the Cyclin-Dependent Kinase Inhibitor P21 in Pancreatic CarcinomaAmerican journal of Pathology1471995Search in Google Scholar
Dardare, J., Witz, A., Merlin, J. L., Gilson, P. & Harlé, A. SMAD4 and the TGFB pathway in patients with pancreatic ductal adenocarcinoma. International Journal of Molecular Sciences vol. 21 Preprint at https://doi.org/10.3390/ijms21103534 (2020)DardareJ.WitzA.MerlinJ. L.GilsonP.HarléA.SMAD4 and the TGFB pathway in patients with pancreatic ductal adenocarcinomaInternational Journal of Molecular Sciences21Preprint at https://doi.org/10.3390/ijms211035342020Search in Google Scholar
Bartsch, D. K. et al. CDKN2A Germline Mutations in Familial Pancreatic Cancer. (2002) doi:10.1097/01.SLA0000036393.89509.4EBartschD. K.CDKN2A Germline Mutations in Familial Pancreatic Cancer200210.1097/01.SLA0000036393.89509.4EOpen DOISearch in Google Scholar
Lomberk, G. et al. Distinct epigenetic landscapes underlie the pathobiology of pancreatic cancer subtypes. Nat Commun9, (2018)LomberkG.Distinct epigenetic landscapes underlie the pathobiology of pancreatic cancer subtypesNat Commun92018Search in Google Scholar
Hayashi, A., Hong, J. & Iacobuzio-Donahue, C. A. The pancreatic cancer genome revisited. Nat Rev Gastroenterol Hepatol18, 469–481 (2021)HayashiA.HongJ.Iacobuzio-DonahueC. A.The pancreatic cancer genome revisitedNat Rev Gastroenterol Hepatol184694812021Search in Google Scholar
Montalto, F. I. & De Amicis, F. Cyclin D1 in Cancer: A Molecular Connection for Cell Cycle Control, Adhesion and Invasion in Tumor and Stroma. Cells vol. 9 Preprint at https://doi.org/10.3390/cells9122648 (2020)MontaltoF. I.De AmicisF.Cyclin D1 in Cancer: A Molecular Connection for Cell Cycle Control, Adhesion and Invasion in Tumor and StromaCells9Preprint at https://doi.org/10.3390/cells91226482020Search in Google Scholar
Goodwin, C. M. et al. Combination Therapies with CDK4/6 Inhibitors to Treat KRAS-Mutant Pancreatic Cancer. Cancer Res83, 141–157 (2023)GoodwinC. M.Combination Therapies with CDK4/6 Inhibitors to Treat KRAS-Mutant Pancreatic CancerCancer Res831411572023Search in Google Scholar
Jenke, R., Reßing, N., Hansen, F. K., Aigner, A. & Büch, T. cancers Anticancer Therapy with HDAC Inhibitors: Mechanism-Based Combination Strategies and Future Perspectives. Cancers (Basel)13, 634 (2021)JenkeR.ReßingN.HansenF. K.AignerA.BüchT.cancers Anticancer Therapy with HDAC Inhibitors: Mechanism-Based Combination Strategies and Future PerspectivesCancers (Basel)136342021Search in Google Scholar
Licht, J. D. & Bennett, R. L. Leveraging epigenetics to enhance the efficacy of immunotherapy. Clinical Epigenetics vol. 13 Preprint at https://doi.org/10.1186/s13148-021-01100-x (2021)LichtJ. D.BennettR. L.Leveraging epigenetics to enhance the efficacy of immunotherapyClinical Epigenetics13Preprint at https://doi.org/10.1186/s13148-021-01100-x2021Search in Google Scholar
Knoche, S. M. et al. The histone deacetylase inhibitor M344 as a multifaceted therapy for pancreatic cancer. PLoS One17, (2022)KnocheS. M.The histone deacetylase inhibitor M344 as a multifaceted therapy for pancreatic cancerPLoS One172022Search in Google Scholar
Simões-Pires, C. et al. HDAC6 as a target for neurodegenerative diseases: What makes it different from the other HDACs? Molecular Neurodegeneration vol. 8 Preprint at https://doi.org/10.1186/1750-1326-8-7 (2013)Simões-PiresC.HDAC6 as a target for neurodegenerative diseases: What makes it different from the other HDACs?Molecular Neurodegeneration8Preprint at https://doi.org/10.1186/1750-1326-8-72013Search in Google Scholar
van Kampen, J. G. M. et al. Epigenetic targeting in pancreatic cancer. Cancer Treat Rev40, 656–664 (2014)van KampenJ. G. M.Epigenetic targeting in pancreatic cancerCancer Treat Rev406566642014Search in Google Scholar
Goto, K. et al. Novel chemoimmunotherapeutic strategy for hepatocellular carcinoma based on a genome-wide association study. Sci Rep6, 38407 (2016)GotoK.Novel chemoimmunotherapeutic strategy for hepatocellular carcinoma based on a genome-wide association studySci Rep6384072016Search in Google Scholar
Sun, T. et al. Histone deacetylase inhibition up-regulates MHC class I to facilitate cytotoxic T lymphocyte-mediated tumor cell killing in glioma cells. J Cancer10, 5638–5645 (2019)SunT.Histone deacetylase inhibition up-regulates MHC class I to facilitate cytotoxic T lymphocyte-mediated tumor cell killing in glioma cellsJ Cancer10563856452019Search in Google Scholar
Ritter, C. et al. Reversal of epigenetic silencing of MHC class I chain-related protein A and B improves immune recognition of Merkel cell carcinoma. Sci Rep6, 21678 (2016)RitterC.Reversal of epigenetic silencing of MHC class I chain-related protein A and B improves immune recognition of Merkel cell carcinomaSci Rep6216782016Search in Google Scholar
Ugurel, S. et al. MHC class-I downregulation in PD-1/PD-L1 inhibitor refractory Merkel cell carcinoma and its potential reversal by histone deacetylase inhibition: a case series. Cancer Immunology, Immunotherapy68, 983–990 (2019)UgurelS.MHC class-I downregulation in PD-1/PD-L1 inhibitor refractory Merkel cell carcinoma and its potential reversal by histone deacetylase inhibition: a case seriesCancer Immunology, Immunotherapy689839902019Search in Google Scholar
Mustafa, A.-H. M. & Krämer, O. H. Pharmacological Modulation of the Crosstalk between Aberrant Janus Kinase Signaling and Epigenetic Modifiers of the Histone Deacetylase Family to Treat Cancer. Pharmacol Rev75, 35 (2023)MustafaA.-H. M.KrämerO. H.Pharmacological Modulation of the Crosstalk between Aberrant Janus Kinase Signaling and Epigenetic Modifiers of the Histone Deacetylase Family to Treat CancerPharmacol Rev75352023Search in Google Scholar
Ashry, R. et al. NOXA Accentuates Apoptosis Induction by a Novel Histone Deacetylase Inhibitor. Cancers (Basel)15, (2023)AshryR.NOXA Accentuates Apoptosis Induction by a Novel Histone Deacetylase InhibitorCancers (Basel)152023Search in Google Scholar
Okano, M., Bell, D. W., Haber, D. A. & Li, E. DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian Development. Cell99, 247–257 (1999)OkanoM.BellD. W.HaberD. A.LiE.DNA Methyltransferases Dnmt3a and Dnmt3b Are Essential for De Novo Methylation and Mammalian DevelopmentCell992472571999Search in Google Scholar
Scourzic, L., Mouly, E. & Bernard, O. A. TET proteins and the control of cytosine demethylation in cancer. Genome Med7, (2015)ScourzicL.MoulyE.BernardO. A.TET proteins and the control of cytosine demethylation in cancerGenome Med72015Search in Google Scholar
Jones, P. A. & Baylin, S. B. The fundamental role of epigenetic events in cancer. Nat Rev Genet3, 415–428 (2002)JonesP. A.BaylinS. B.The fundamental role of epigenetic events in cancerNat Rev Genet34154282002Search in Google Scholar
Jeziorska, D. M. et al. DNA methylation of intragenic CpG islands depends on their transcriptional activity during differentiation and disease. Proc Natl Acad Sci U S A114, E7526–E7535 (2017)JeziorskaD. M.DNA methylation of intragenic CpG islands depends on their transcriptional activity during differentiation and diseaseProc Natl Acad Sci U S A114E7526E75352017Search in Google Scholar
Lander, E. S. et al. Initial sequencing and analysis of the human genome. Nature409, 860–921 (2001)LanderE. S.Initial sequencing and analysis of the human genomeNature4098609212001Search in Google Scholar
Herman, J. G. & Baylin, S. B. Gene Silencing in Cancer in Association with Promoter Hypermethylation. New England Journal of Medicine349, 2042–2054 (2003)HermanJ. G.BaylinS. B.Gene Silencing in Cancer in Association with Promoter HypermethylationNew England Journal of Medicine349204220542003Search in Google Scholar
Zhu, J. et al. Integrating genome and methylome data to identify candidate DNA methylation biomarkers for pancreatic cancer risk. Cancer Epidemiology Biomarkers and Prevention30, 2079–2087 (2021)ZhuJ.Integrating genome and methylome data to identify candidate DNA methylation biomarkers for pancreatic cancer riskCancer Epidemiology Biomarkers and Prevention30207920872021Search in Google Scholar
Ozturk, H. et al. ISL2 is a putative tumor suppressor whose epigenetic silencing reprograms the metabolism of pancreatic cancer. Dev Cell57, 1331–1346.e9 (2022)OzturkH.ISL2 is a putative tumor suppressor whose epigenetic silencing reprograms the metabolism of pancreatic cancerDev Cell5713311346.e92022Search in Google Scholar
Sato, N., Matsubayashi, H., Abe, T., Fukushima, N. & Goggins, M. Epigenetic Down-Regulation of CDKN1C/p57KIP2 in Pancreatic Ductal Neoplasms Identified by Gene Expression Profiling. Clinical Cancer Research11, 4681–4688 (2005)SatoN.MatsubayashiH.AbeT.FukushimaN.GogginsM.Epigenetic Down-Regulation of CDKN1C/p57KIP2 in Pancreatic Ductal Neoplasms Identified by Gene Expression ProfilingClinical Cancer Research11468146882005Search in Google Scholar
Eyres, M. et al. TET2 Drives 5hmc Marking of GATA6 and Epigenetically Defines Pancreatic Ductal Adenocarcinoma Transcriptional Subtypes. Gastroenterology161, 653–668.e16 (2021)EyresM.TET2 Drives 5hmc Marking of GATA6 and Epigenetically Defines Pancreatic Ductal Adenocarcinoma Transcriptional SubtypesGastroenterology161653668.e162021Search in Google Scholar
Xiao, Q. et al. Cancer-associated fibroblasts in pancreatic cancer are reprogrammed by tumor-induced alterations in genomic DNA methylation. Cancer Res76, 5395–5404 (2016)XiaoQ.Cancer-associated fibroblasts in pancreatic cancer are reprogrammed by tumor-induced alterations in genomic DNA methylationCancer Res76539554042016Search in Google Scholar
Zhang, M. et al. Pancreatic cancer cells render tumor-associated macrophages metabolically reprogrammed by a GARP and DNA methylation-mediated mechanism. Signal Transduct Target Ther6, (2021)ZhangM.Pancreatic cancer cells render tumor-associated macrophages metabolically reprogrammed by a GARP and DNA methylation-mediated mechanismSignal Transduct Target Ther62021Search in Google Scholar
Tang, R.-Z. et al. DNA methyltransferase 1 and Krüppel-like factor 4 axis regulates macrophage inflammation and atherosclerosis. J Mol Cell Cardiol128, 11–24 (2019)TangR.-Z.DNA methyltransferase 1 and Krüppel-like factor 4 axis regulates macrophage inflammation and atherosclerosisJ Mol Cell Cardiol12811242019Search in Google Scholar
Mees, S. T. et al. EP300—A miRNA-regulated metastasis suppressor gene in ductal adenocarcinomas of the pancreas. Int J Cancer126, 114–124 (2010)MeesS. T.EP300—A miRNA-regulated metastasis suppressor gene in ductal adenocarcinomas of the pancreasInt J Cancer1261141242010Search in Google Scholar
Cai, M. H. et al. Depletion of HDAC1, 7 and 8 by Histone Deacetylase Inhibition Confers Elimination of Pancreatic Cancer Stem Cells in Combination with Gemcitabine. Sci Rep8, (2018)CaiM. H.Depletion of HDAC1, 7 and 8 by Histone Deacetylase Inhibition Confers Elimination of Pancreatic Cancer Stem Cells in Combination with GemcitabineSci Rep82018Search in Google Scholar
Klieser, E. et al. Role of histone deacetylases in pancreas: Implications for pathogenesis and therapy. World J Gastrointest Oncol7, 473–483 (2015)KlieserE.Role of histone deacetylases in pancreas: Implications for pathogenesis and therapyWorld J Gastrointest Oncol74734832015Search in Google Scholar
Brand, M. et al. Small Molecule Inhibitors of Bromodomain–Acetyl-lysine Interactions. ACS Chem Biol10, 22–39 (2015)BrandM.Small Molecule Inhibitors of Bromodomain–Acetyl-lysine InteractionsACS Chem Biol1022392015Search in Google Scholar
Donati, B., Lorenzini, E. & Ciarrocchi, A. BRD4 and Cancer: Going beyond transcriptional regulation. Molecular Cancer vol. 17 Preprint at https://doi.org/10.1186/s12943-018-0915-9 (2018)DonatiB.LorenziniE.CiarrocchiA.BRD4 and Cancer: Going beyond transcriptional regulationMolecular Cancer17Preprint at https://doi.org/10.1186/s12943-018-0915-92018Search in Google Scholar
Shi, J. & Vakoc, C. R. The Mechanisms behind the Therapeutic Activity of BET Bromodomain Inhibition. Molecular Cell vol. 54 728–736 Preprint at https://doi.org/10.1016/j.molcel.2014.05.016 (2014)ShiJ.VakocC. R.The Mechanisms behind the Therapeutic Activity of BET Bromodomain InhibitionMolecular Cell54728736Preprint at https://doi.org/10.1016/j.molcel.2014.05.0162014Search in Google Scholar
Greer, E. L. & Shi, Y. Histone methylation: A dynamic mark in health, disease and inheritance. Nature Reviews Genetics vol. 13 343–357 Preprint at https://doi.org/10.1038/nrg3173 (2012)GreerE. L.ShiY.Histone methylation: A dynamic mark in health, disease and inheritanceNature Reviews Genetics13343357Preprint at https://doi.org/10.1038/nrg31732012Search in Google Scholar
Chen, Y. et al. The role of histone methylation in the development of digestive cancers: a potential direction for cancer management. Signal Transduction and Targeted Therapy vol. 5 Preprint at https://doi.org/10.1038/s41392-020-00252-1 (2020)ChenY.The role of histone methylation in the development of digestive cancers: a potential direction for cancer managementSignal Transduction and Targeted Therapy5Preprint at https://doi.org/10.1038/s41392-020-00252-12020Search in Google Scholar
Benitz, S. et al. Ring1b-dependent epigenetic remodelling is an essential prerequisite for pancreatic carcinogenesis. Gut68, 2007 (2019)BenitzS.Ring1b-dependent epigenetic remodelling is an essential prerequisite for pancreatic carcinogenesisGut6820072019Search in Google Scholar
Andricovich, J. et al. Loss of KDM6A Activates Super-Enhancers to Induce Gender-Specific Squamous-like Pancreatic Cancer and Confers Sensitivity to BET Inhibitors. Cancer Cell33, 512–526.e8 (2018)AndricovichJ.Loss of KDM6A Activates Super-Enhancers to Induce Gender-Specific Squamous-like Pancreatic Cancer and Confers Sensitivity to BET InhibitorsCancer Cell33512526.e82018Search in Google Scholar
Rao, R. A. et al. Ezh2 mediated H3K27me3 activity facilitates somatic transition during human pluripotent reprogramming. Sci Rep5, (2015)RaoR. A.Ezh2 mediated H3K27me3 activity facilitates somatic transition during human pluripotent reprogrammingSci Rep52015Search in Google Scholar
Ougolkov, A. V., Bilim, V. N. & Billadeau, D. D. Regulation of pancreatic tumor cell proliferation and chemoresistance by the histone methyltransferase enhancer of zeste homologue 2. Clinical Cancer Research14, 6790–6796 (2008)OugolkovA. V.BilimV. N.BilladeauD. D.Regulation of pancreatic tumor cell proliferation and chemoresistance by the histone methyltransferase enhancer of zeste homologue 2Clinical Cancer Research14679067962008Search in Google Scholar
Toll, A. D. et al. Implications of enhancer of zeste homologue 2 expression in pancreatic ductal adenocarcinoma. Hum Pathol41, 1205–1209 (2010)TollA. D.Implications of enhancer of zeste homologue 2 expression in pancreatic ductal adenocarcinomaHum Pathol41120512092010Search in Google Scholar
Han, T. et al. EZH2 Promotes Cell Migration and Invasion but Not Alters Cell Proliferation by Suppressing E-Cadherin, Partly through Association with MALAT-1 in Pancreatic Cancer. Oncotarget vol. 7 www.impactjournals.com/oncotarget/HanT.EZH2 Promotes Cell Migration and Invasion but Not Alters Cell Proliferation by Suppressing E-Cadherin, Partly through Association with MALAT-1 in Pancreatic CancerOncotarget7www.impactjournals.com/oncotarget/Search in Google Scholar
Aghdassi, A. et al. Recruitment of histone deacetylases HDAC1 and HDAC2 by the transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancer. Gut61, 439 (2012)AghdassiA.Recruitment of histone deacetylases HDAC1 and HDAC2 by the transcriptional repressor ZEB1 downregulates E-cadherin expression in pancreatic cancerGut614392012Search in Google Scholar
Song, Y., Washington, M. K. & Crawford, H. C. Loss of FOXA1/2 is essential for the epithelial-to-mesenchymal transition in pancreatic cancer. Cancer Res70, 2115–2125 (2010)SongY.WashingtonM. K.CrawfordH. C.Loss of FOXA1/2 is essential for the epithelial-to-mesenchymal transition in pancreatic cancerCancer Res70211521252010Search in Google Scholar
Roe, J. S. et al. Enhancer Reprogramming Promotes Pancreatic Cancer Metastasis. Cell170, 875–888.e20 (2017)RoeJ. S.Enhancer Reprogramming Promotes Pancreatic Cancer MetastasisCell170875888.e202017Search in Google Scholar
Hessmann, E., Johnsen, S. A., Siveke, J. T. & Ellenrieder, V. Epigenetic treatment of pancreatic cancer: Is there a therapeutic perspective on the horizon? Gut66, 168–179 (2017)HessmannE.JohnsenS. A.SivekeJ. T.EllenriederV.Epigenetic treatment of pancreatic cancer: Is there a therapeutic perspective on the horizon?Gut661681792017Search in Google Scholar
Roca, M. S. et al. HDAC class I inhibitor domatinostat sensitizes pancreatic cancer to chemotherapy by targeting cancer stem cell compartment via FOXM1 modulation. Journal of Experimental and Clinical Cancer Research41, (2022)RocaM. S.HDAC class I inhibitor domatinostat sensitizes pancreatic cancer to chemotherapy by targeting cancer stem cell compartment via FOXM1 modulationJournal of Experimental and Clinical Cancer Research412022Search in Google Scholar
Romeo, M. A. et al. Acetylation increases expression, interaction with TRAPPC4 and surface localization of PD-L1. Discover Oncology14, (2023)RomeoM. A.Acetylation increases expression, interaction with TRAPPC4 and surface localization of PD-L1Discover Oncology142023Search in Google Scholar
Qu, L. et al. The nuclear transportation of PD-L1 and the function in tumor immunity and progression. Cancer Immunology, Immunotherapy71, 2313–2323 (2022)QuL.The nuclear transportation of PD-L1 and the function in tumor immunity and progressionCancer Immunology, Immunotherapy71231323232022Search in Google Scholar
Xiong, W., Gao, Y., Wei, W. & Zhang, J. Extracellular and nuclear PD-L1 in modulating cancer immunotherapy. Trends Cancer7, 837–846 (2021)XiongW.GaoY.WeiW.ZhangJ.Extracellular and nuclear PD-L1 in modulating cancer immunotherapyTrends Cancer78378462021Search in Google Scholar
Zeng, Y. et al. MARK2 regulates chemotherapeutic responses through class IIa HDAC-YAP axis in pancreatic cancer. Oncogene41, 3859–3875 (2022)ZengY.MARK2 regulates chemotherapeutic responses through class IIa HDAC-YAP axis in pancreatic cancerOncogene41385938752022Search in Google Scholar
Kuo, T. L., Cheng, K. H., Chen, L. T. & Hung, W. C. ARID1A loss in pancreas leads to islet developmental defect and metabolic disturbance. iScience26, (2023)KuoT. L.ChengK. H.ChenL. T.HungW. C.ARID1A loss in pancreas leads to islet developmental defect and metabolic disturbanceiScience262023Search in Google Scholar
Russell, M. A. et al. HLA Class II Antigen Processing and Presentation Pathway Components Demonstrated by Transcriptome and Protein Analyses of Islet β-Cells From Donors With Type 1 Diabetes. Diabetes68, 988–1001 (2019)RussellM. A.HLA Class II Antigen Processing and Presentation Pathway Components Demonstrated by Transcriptome and Protein Analyses of Islet β-Cells From Donors With Type 1 DiabetesDiabetes6898810012019Search in Google Scholar
Sixto-López, Y. et al. Hydroxamic acid derivatives as HDAC1, HDAC6 and HDAC8 inhibitors with antiproliferative activity in cancer cell lines. Sci Rep10, (2020)Sixto-LópezY.Hydroxamic acid derivatives as HDAC1, HDAC6 and HDAC8 inhibitors with antiproliferative activity in cancer cell linesSci Rep102020Search in Google Scholar
Butler, K. V. et al. Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin A. J Am Chem Soc132, 10842–10846 (2010)ButlerK. V.Rational design and simple chemistry yield a superior, neuroprotective HDAC6 inhibitor, tubastatin AJ Am Chem Soc13210842108462010Search in Google Scholar
Kong, Y. et al. Histone deacetylase cytoplasmic trapping by a novel fluorescent HDAC inhibitor. Mol Cancer Ther10, 1591–1599 (2011)KongY.Histone deacetylase cytoplasmic trapping by a novel fluorescent HDAC inhibitorMol Cancer Ther10159115992011Search in Google Scholar
Estiu, G. et al. On the inhibition of histone deacetylase 8. Bioorg Med Chem18, 4103–4110 (2010)EstiuG.On the inhibition of histone deacetylase 8Bioorg Med Chem18410341102010Search in Google Scholar
Charrier, C. et al. Synthesis and Modeling of New Benzofuranone Histone Deacetylase Inhibitors that Stimulate Tumor Suppressor Gene Expression. J Med Chem52, 3112–3115 (2009)CharrierC.Synthesis and Modeling of New Benzofuranone Histone Deacetylase Inhibitors that Stimulate Tumor Suppressor Gene ExpressionJ Med Chem52311231152009Search in Google Scholar
Khochbin, S., Verdel, A., Lemercier, C. & Seigneurin-Berny, D. Functional significance of histone deacetylase diversity. Curr Opin Genet Dev11, 162–166 (2001)KhochbinS.VerdelA.LemercierC.Seigneurin-BernyD.Functional significance of histone deacetylase diversityCurr Opin Genet Dev111621662001Search in Google Scholar
Wang, D.-F., Helquist, P., Wiech, N. L. & Wiest, O. Toward Selective Histone Deacetylase Inhibitor Design: Homology Modeling, Docking Studies, and Molecular Dynamics Simulations of Human Class I Histone Deacetylases. J Med Chem48, 6936–6947 (2005)WangD.-F.HelquistP.WiechN. L.WiestO.Toward Selective Histone Deacetylase Inhibitor Design: Homology Modeling, Docking Studies, and Molecular Dynamics Simulations of Human Class I Histone DeacetylasesJ Med Chem48693669472005Search in Google Scholar
Mazur, P. K. et al. Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinoma. Nat Med21, 1163–1171 (2015)MazurP. K.Combined inhibition of BET family proteins and histone deacetylases as a potential epigenetics-based therapy for pancreatic ductal adenocarcinomaNat Med21116311712015Search in Google Scholar
Maietta, I. et al. Synergistic Antitumoral Effect of Epigenetic Inhibitors and Gemcitabine in Pancreatic Cancer Cells. Pharmaceuticals15, (2022)MaiettaI.Synergistic Antitumoral Effect of Epigenetic Inhibitors and Gemcitabine in Pancreatic Cancer CellsPharmaceuticals152022Search in Google Scholar
Bai, J., Demirjian, A., Sui, J., Marasco, W. & Callery, M. P. Histone deacetylase inhibitor trichostatin A and proteasome inhibitor PS-341 synergistically induce apoptosis in pancreatic cancer cells. Biochem Biophys Res Commun348, 1245–1253 (2006)BaiJ.DemirjianA.SuiJ.MarascoW.CalleryM. P.Histone deacetylase inhibitor trichostatin A and proteasome inhibitor PS-341 synergistically induce apoptosis in pancreatic cancer cellsBiochem Biophys Res Commun348124512532006Search in Google Scholar
Christmas, B. J. et al. Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCs. Cancer Immunol Res6, 1561–1577 (2018)ChristmasB. J.Entinostat converts immune-resistant breast and pancreatic cancers into checkpoint-responsive tumors by reprogramming tumor-infiltrating MDSCsCancer Immunol Res6156115772018Search in Google Scholar
Poklepovic, A. S. et al. A phase 1 study of neoadjuvant chemotherapy followed by concurrent chemoradiation with gemcitabine, sorafenib, and vorinostat in pancreatic cancer. Journal of Clinical Oncology39, e16268–e16268 (2021)PoklepovicA. S.A phase 1 study of neoadjuvant chemotherapy followed by concurrent chemoradiation with gemcitabine, sorafenib, and vorinostat in pancreatic cancerJournal of Clinical Oncology39e16268e162682021Search in Google Scholar
Streubel, G. et al. Histone deacetylase inhibitor resminostat in combination with sorafenib counteracts platelet-mediated pro-tumoral effects in hepatocellular carcinoma. Sci Rep11, (2021)StreubelG.Histone deacetylase inhibitor resminostat in combination with sorafenib counteracts platelet-mediated pro-tumoral effects in hepatocellular carcinomaSci Rep112021Search in Google Scholar
Wang, Z. et al. SETD5-Coordinated Chromatin Reprogramming Regulates Adaptive Resistance to Targeted Pancreatic Cancer Therapy. Cancer Cell37, 834–849.e13 (2020)WangZ.SETD5-Coordinated Chromatin Reprogramming Regulates Adaptive Resistance to Targeted Pancreatic Cancer TherapyCancer Cell37834849.e132020Search in Google Scholar
Chan, E. et al. Phase i trial of vorinostat added to chemoradiation with capecitabine in pancreatic cancer. Radiotherapy and Oncology119, 312–318 (2016)ChanE.Phase i trial of vorinostat added to chemoradiation with capecitabine in pancreatic cancerRadiotherapy and Oncology1193123182016Search in Google Scholar
Ubbert, M. L. ¨ et al. MD 12,13 ; Hans-Walter Lindemann, MD 14 ; Carsten Müller-Tidow, MD 5,15,16 ; Gerhard Heil. J Clin Oncol17, 257–270Ubbert, M. L. ¨ et al. MD 12,13 ; Hans-Walter Lindemann, MD 14 ; Carsten Müller-Tidow, MD 5,15,16 ; Gerhard Heil.J Clin Oncol17257270Search in Google Scholar
Meier, R. et al. The antileukemic activity of decitabine upon PML/RARA-negative AML blasts is supported by all-trans retinoic acid: in vitro and in vivo evidence for cooperation. Blood Cancer J12, (2022)MeierR.The antileukemic activity of decitabine upon PML/RARA-negative AML blasts is supported by all-trans retinoic acid: in vitro and in vivo evidence for cooperationBlood Cancer J122022Search in Google Scholar
Li, Y. et al. Alterations of specific chromatin conformation affect ATRA-induced leukemia cell differentiation. Cell Death Dis9, (2018)LiY.Alterations of specific chromatin conformation affect ATRA-induced leukemia cell differentiationCell Death Dis92018Search in Google Scholar
Trus, M. R. et al. The histone deacetylase inhibitor valproic acid alters sensitivity towards all trans retinoic acid in acute myeloblastic leukemia cells. Leukemia19, 1161–1168 (2005)TrusM. R.The histone deacetylase inhibitor valproic acid alters sensitivity towards all trans retinoic acid in acute myeloblastic leukemia cellsLeukemia19116111682005Search in Google Scholar
Luu, T. et al. Phase I trial of belinostat in combination with 13-cis-retinoic acid in advanced solid tumor malignancies: a California Cancer Consortium NCI/CTEP sponsored trial. Cancer Chemother Pharmacol84, 1201–1208 (2019)LuuT.Phase I trial of belinostat in combination with 13-cis-retinoic acid in advanced solid tumor malignancies: a California Cancer Consortium NCI/CTEP sponsored trialCancer Chemother Pharmacol84120112082019Search in Google Scholar
Neureiter, D. et al. Apoptosis, proliferation and differentiation patterns are influenced by Zebularine and SAHA in pancreatic cancer models. Scand J Gastroenterol42, 103–116 (2007)NeureiterD.Apoptosis, proliferation and differentiation patterns are influenced by Zebularine and SAHA in pancreatic cancer modelsScand J Gastroenterol421031162007Search in Google Scholar
Guo, Z. et al. Design and Synthesis of Dual EZH2/BRD4 Inhibitors to Target Solid Tumors. J Med Chem65, 6573–6592 (2022)GuoZ.Design and Synthesis of Dual EZH2/BRD4 Inhibitors to Target Solid TumorsJ Med Chem65657365922022Search in Google Scholar
He, S. et al. Potent Dual BET/HDAC Inhibitors for Efficient Treatment of Pancreatic Cancer. Angewandte Chemie International Edition59, 3028–3032 (2020)HeS.Potent Dual BET/HDAC Inhibitors for Efficient Treatment of Pancreatic CancerAngewandte Chemie International Edition59302830322020Search in Google Scholar
Lassen, U. et al. A phase i study of the safety and pharmacokinetics of the histone deacetylase inhibitor belinostat administered in combination with carboplatin and/or paclitaxel in patients with solid tumours. Br J Cancer103, 12–17 (2010)LassenU.A phase i study of the safety and pharmacokinetics of the histone deacetylase inhibitor belinostat administered in combination with carboplatin and/or paclitaxel in patients with solid tumoursBr J Cancer10312172010Search in Google Scholar
Ikeda, M. et al. Phase I study of resminostat, an HDAC inhibitor, combined with S-1 in patients with pre-treated biliary tract or pancreatic cancer. Invest New Drugs37, 109–117 (2019)IkedaM.Phase I study of resminostat, an HDAC inhibitor, combined with S-1 in patients with pre-treated biliary tract or pancreatic cancerInvest New Drugs371091172019Search in Google Scholar