Phase I/II sequencing study of azacitidine, epacadostat, and pembrolizumab in advanced solid tumors

[1]  P. Trzonkowski,et al.  Regulatory T Cells-Related Genes Are under DNA Methylation Influence , 2021, International journal of molecular sciences.

[2]  Chenguang Wang,et al.  Phase I Study of Entinostat and Nivolumab with or without Ipilimumab in Advanced Solid Tumors (ETCTN-9844) , 2021, Clinical Cancer Research.

[3]  Ronen Levy,et al.  Anti-tumour immunity induces aberrant peptide presentation in melanoma , 2020, Nature.

[4]  L. Leopold,et al.  28 Retrospective pooled analysis of epacadostat clinical studies identifies doses required for maximal pharmacodynamic effect in anti-PD-1 combination studies , 2020, Journal for ImmunoTherapy of Cancer.

[5]  Z. Ronai,et al.  PRMT5 control of cGAS/STING and NLRC5 pathways defines melanoma response to antitumor immunity , 2020, Science Translational Medicine.

[6]  Aibin He,et al.  Distinct epigenetic features of tumor-reactive CD8+ T cells in colorectal cancer patients revealed by genome-wide DNA methylation analysis , 2019, Genome Biology.

[7]  T. Tzai,et al.  Indoleamine-2,3-dioxygenase-1 expression predicts poorer survival and up-regulates ZEB2 expression in human early stage bladder cancer. , 2019, Urologic oncology.

[8]  D. Greenawalt,et al.  Interferon ɣ (IFN-ɣ) gene signature and tryptophan 2,3-dioxygenase 2 (TDO2) gene expression: a potential predictive composite biomarker for linrodostat mesylate (BMS-986205; indoleamine 2,3-dioxygenase 1 inhibitor [IDO1i]) + nivolumab (NIVO) , 2019, Annals of Oncology.

[9]  S. Baylin,et al.  The emerging role of epigenetic therapeutics in immuno-oncology , 2019, Nature Reviews Clinical Oncology.

[10]  James R. Anderson,et al.  Epacadostat plus pembrolizumab versus placebo plus pembrolizumab in patients with unresectable or metastatic melanoma (ECHO-301/KEYNOTE-252): a phase 3, randomised, double-blind study. , 2019, The Lancet. Oncology.

[11]  G. Gibney,et al.  Phase 1/2 study of epacadostat in combination with ipilimumab in patients with unresectable or metastatic melanoma , 2019, Journal of Immunotherapy for Cancer.

[12]  J. Winter,et al.  Immune Profiling and Quantitative Analysis Decipher the Clinical Role of Immune-Checkpoint Expression in the Tumor Immune Microenvironment of DLBCL , 2019, Cancer Immunology Research.

[13]  Y. Maehara,et al.  Co-expression of IDO1 and PD-L1 in lung squamous cell carcinoma: Potential targets of novel combination therapy. , 2019, Lung cancer.

[14]  C. Zahnow,et al.  Targeting CDK9 Reactivates Epigenetically Silenced Genes in Cancer , 2018, Cell.

[15]  J. Luke,et al.  Reimagining IDO Pathway Inhibition in Cancer Immunotherapy via Downstream Focus on the Tryptophan–Kynurenine–Aryl Hydrocarbon Axis , 2018, Clinical Cancer Research.

[16]  J. Lunceford,et al.  Pan-tumor genomic biomarkers for PD-1 checkpoint blockade–based immunotherapy , 2018, Science.

[17]  David C. Smith,et al.  Epacadostat Plus Pembrolizumab in Patients With Advanced Solid Tumors: Phase I Results From a Multicenter, Open-Label Phase I/II Trial (ECHO-202/KEYNOTE-037) , 2018, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[18]  J. Luke,et al.  T Cell–Inflamed versus Non-T Cell–Inflamed Tumors: A Conceptual Framework for Cancer Immunotherapy Drug Development and Combination Therapy Selection , 2018, Cancer Immunology Research.

[19]  G. Freeman,et al.  LSD1 Ablation Stimulates Anti-tumor Immunity and Enables Checkpoint Blockade , 2018, Cell.

[20]  A. Daud,et al.  Epacadostat plus nivolumab for advanced melanoma: Updated phase 2 results of the ECHO-204 study. , 2018 .

[21]  M. Baretti,et al.  The role of epigenetic therapies in colorectal cancer. , 2018, Current problems in cancer.

[22]  D. Wainwright,et al.  IDO1 in cancer: a Gemini of immune checkpoints , 2018, Cellular & Molecular Immunology.

[23]  M. Mino‐Kenudson,et al.  PD-L1 and IDO1 Are Expressed in Poorly Differentiated Thyroid Carcinoma , 2018, Endocrine Pathology.

[24]  Jeremy Stinson,et al.  Silencing of retrotransposons by SETDB1 inhibits the interferon response in acute myeloid leukemia , 2017, The Journal of cell biology.

[25]  I. Durán,et al.  Biomarkers of response to PD-1/PD-L1 inhibition. , 2017, Critical reviews in oncology/hematology.

[26]  David C. Smith,et al.  Epacadostat plus pembrolizumab in patients with advanced urothelial carcinoma: Preliminary phase I/II results of ECHO-202/KEYNOTE-037. , 2017 .

[27]  M. Bower,et al.  INCB24360 (Epacadostat), a Highly Potent and Selective Indoleamine-2,3-dioxygenase 1 (IDO1) Inhibitor for Immuno-oncology , 2017, ACS medicinal chemistry letters.

[28]  H. Koblish,et al.  Abstract 4904: The BET inhibitor INCB054329 enhances the activity of checkpoint modulation in syngeneic tumor models , 2016 .

[29]  C. Zahnow,et al.  Combining Epigenetic and Immunotherapy to Combat Cancer. , 2016, Cancer research.

[30]  Nita Ahuja,et al.  Epigenetic Therapeutics: A New Weapon in the War Against Cancer. , 2016, Annual review of medicine.

[31]  M. Beckmann,et al.  Abstract B32: Inhibiting DNA methylation causes an interferon response in cancer via dsRNA including endogenous retroviruses , 2016 .

[32]  Trevor J Pugh,et al.  DNA-Demethylating Agents Target Colorectal Cancer Cells by Inducing Viral Mimicry by Endogenous Transcripts , 2015, Cell.

[33]  R. Emerson,et al.  PD-1 blockade induces responses by inhibiting adaptive immune resistance , 2014, Nature.

[34]  H. Koblish,et al.  Mechanism of tumor rejection with doublets of CTLA-4, PD-1/PD-L1, or IDO blockade involves restored IL-2 production and proliferation of CD8+ T cells directly within the tumor microenvironment , 2014, Journal of Immunotherapy for Cancer.

[35]  Peter A. Jones,et al.  Alterations of immune response of non-small cell lung cancer with Azacytidine , 2013, Oncotarget.

[36]  Jason B. Williams,et al.  Up-Regulation of PD-L1, IDO, and Tregs in the Melanoma Tumor Microenvironment Is Driven by CD8+ T Cells , 2013, Science Translational Medicine.

[37]  J. Wolchok,et al.  Indoleamine 2,3-dioxygenase is a critical resistance mechanism in antitumor T cell immunotherapy targeting CTLA-4 , 2013, The Journal of experimental medicine.

[38]  S. Quezada,et al.  Exploiting CTLA-4, PD-1 and PD-L1 to reactivate the host immune response against cancer , 2013, British Journal of Cancer.

[39]  P. Vyas,et al.  Azacitidine augments expansion of regulatory T cells after allogeneic stem cell transplantation in patients with acute myeloid leukemia (AML). , 2012, Blood.

[40]  J. Bluestone,et al.  Control of peripheral T‐cell tolerance and autoimmunity via the CTLA‐4 and PD‐1 pathways , 2008, Immunological reviews.

[41]  H. Kajiyama,et al.  Indoleamine 2,3-dioxygenase is a novel prognostic indicator for endometrial cancer , 2006, British Journal of Cancer.

[42]  U. Grohmann,et al.  The Combined Effects of Tryptophan Starvation and Tryptophan Catabolites Down-Regulate T Cell Receptor ζ-Chain and Induce a Regulatory Phenotype in Naive T Cells1 , 2006, The Journal of Immunology.

[43]  N. Yanaihara,et al.  Indoleamine 2,3-Dioxygenase Serves as a Marker of Poor Prognosis in Gene Expression Profiles of Serous Ovarian Cancer Cells , 2005, Clinical Cancer Research.

[44]  B. Baban,et al.  Cutting Edge: Induced Indoleamine 2,3 Dioxygenase Expression in Dendritic Cell Subsets Suppresses T Cell Clonal Expansion1 , 2003, The Journal of Immunology.

[45]  D. Munn,et al.  Tryptophan deprivation sensitizes activated T cells to apoptosis prior to cell division , 2002, Immunology.

[46]  Milton W. Taylor,et al.  Relationship between interferon‐γ, indoleamine 2,3‐dioxygenase, and tryptophan catabolism , 1991, FASEB journal : official publication of the Federation of American Societies for Experimental Biology.

[47]  J. Lunceford,et al.  IFN- γ –related mRNA profile predicts clinical response to PD-1 blockade , 2017 .

[48]  L. Schwartz,et al.  New response evaluation criteria in solid tumours: revised RECIST guideline (version 1.1). , 2009, European journal of cancer.

[49]  Wellbutrin,et al.  Prescribing Information , 2015, European journal of haematology.