Efficacy of azacitidine is independent of molecular and clinical characteristics - an analysis of 128 patients with myelodysplastic syndromes or acute myeloid leukemia and a review of the literature

Azacitidine is the first drug to demonstrate a survival benefit for patients with MDS. However, only half of patients respond and almost all patients eventually relapse. Limited and conflicting data are available on predictive factors influencing response. We analyzed 128 patients from two institutions with MDS or AML treated with azacitidine to identify prognostic indicators. Genetic mutations in ASXL1, RUNX1, DNMT3A, IDH1, IDH2, TET2, TP53, NRAS, KRAS, FLT3, KMT2A-PTD, EZH2, SF3B1, and SRSF2 were assessed by next-generation sequencing. With a median follow up of 5.6 years median survival was 1.3 years with a response rate of 49%. The only variable with significant influence on response was del(20q). All 6 patients responded (p = 0.012) but survival was not improved. No other clinical, cytogenetic or molecular marker for response or survival was identified. Interestingly, patients from poor-risk groups as high-risk cytogenetics (55%), t-MDS/AML (54%), TP53 mutated (48%) or relapsed after chemotherapy (60%) showed a high response rate. Factors associated with shorter survival were low platelets, AML vs. MDS, therapy-related disease, TP53 and KMT2A-PTD. In multivariate analysis anemia, platelets, FLT3-ITD, and therapy-related disease remained in the model. Poor-risk factors such as del(7q)/-7, complex karyotype, ASXL1, RUNX1, EZH2, and TP53 did not show an independent impact. Thus, no clear biomarker for response and survival can be identified. Although a number of publications on predictive markers for response to AZA exist, results are inconsistent and improved response rates did not translate to improved survival. Here, we provide a comprehensive overview comparing the studies published to date.

[1]  S. Nuzhdin,et al.  Suppression of Transposable Elements in Leukemic Stem Cells , 2017, Scientific Reports.

[2]  K. Oetjen,et al.  Immunological effects of hypomethylating agents , 2017, Expert review of hematology.

[3]  R. Fan,et al.  Prognostic significance of TET2 mutations in myelodysplastic syndromes: A meta-analysis. , 2017, Leukemia research.

[4]  Q. Shen,et al.  Demethylating agent decitabine disrupts tumor-induced immune tolerance by depleting myeloid-derived suppressor cells , 2017, Journal of Cancer Research and Clinical Oncology.

[5]  D. Neuberg,et al.  Prognostic Mutations in Myelodysplastic Syndrome after Stem‐Cell Transplantation , 2017, The New England journal of medicine.

[6]  Ming Sun,et al.  Long noncoding RNA ZFAS1 promotes gastric cancer cells proliferation by epigenetically repressing KLF2 and NKD2 expression , 2016, Oncotarget.

[7]  T. Ley,et al.  Decitabine in TP53-Mutated AML. , 2017, New England Journal of Medicine.

[8]  L. Bullinger,et al.  Evaluating the impact of genetic and epigenetic aberrations on survival and response in acute myeloid leukemia patients receiving epigenetic therapy , 2017, Annals of Hematology.

[9]  T. Haferlach,et al.  Application of an NGS‐based 28‐gene panel in myeloproliferative neoplasms reveals distinct mutation patterns in essential thrombocythaemia, primary myelofibrosis and polycythaemia vera , 2016, British journal of haematology.

[10]  P. Walker,et al.  Decitabine Treatment of Glioma-Initiating Cells Enhances Immune Recognition and Killing , 2016, PloS one.

[11]  M. Arcila,et al.  Mutational correlates of response to hypomethylating agent therapy in acute myeloid leukemia , 2016, Haematologica.

[12]  Y. Chung,et al.  Somatic mutations predict outcomes of hypomethylating therapy in patients with myelodysplastic syndrome , 2016, Oncotarget.

[13]  Anne-Kathrin Garz,et al.  A clinical-molecular update on azanucleoside-based therapy for the treatment of hematologic cancers , 2016, Clinical Epigenetics.

[14]  Mario Cazzola,et al.  The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. , 2016, Blood.

[15]  M. Beckmann,et al.  Inhibiting DNA Methylation Causes an Interferon Response in Cancer via dsRNA Including Endogenous Retroviruses , 2016, Cell.

[16]  M. Stratton,et al.  Mutation allele burden remains unchanged in chronic myelomonocytic leukaemia responding to hypomethylating agents , 2016, Nature Communications.

[17]  G. Mufti,et al.  Mutations in histone modulators are associated with prolonged survival during azacitidine therapy , 2015, Oncotarget.

[18]  B. Paluch,et al.  Induction of cancer testis antigen expression in circulating acute myeloid leukemia blasts following hypomethylating agent monotherapy , 2015, Oncotarget.

[19]  T. Haferlach,et al.  Karyotype evolution and acquisition of FLT3 or RAS pathway alterations drive progression of myelodysplastic syndrome to acute myeloid leukemia , 2015, Haematologica.

[20]  L. Ouafik,et al.  Yin and yang of cytidine deaminase roles in clinical response to azacitidine in the elderly: a pharmacogenetics tale. , 2015, Pharmacogenomics.

[21]  E. Hellström-Lindberg,et al.  Imprint of 5-azacytidine on the natural killer cell repertoire during systemic treatment for high-risk myelodysplastic syndrome , 2015, Oncotarget.

[22]  E. Angelucci,et al.  Azacitidine improves the T-cell repertoire in patients with myelodysplastic syndromes and acute myeloid leukemia with multilineage dysplasia. , 2015, Leukemia research.

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

[24]  R. Greil,et al.  International phase 3 study of azacitidine vs conventional care regimens in older patients with newly diagnosed AML with >30% blasts. , 2015, Blood.

[25]  Omar Abdel-Wahab,et al.  Specific molecular signatures predict decitabine response in chronic myelomonocytic leukemia. , 2015, The Journal of clinical investigation.

[26]  Hee-Jin Kim,et al.  Mutations in the Spliceosomal Machinery Genes SRSF2, U2AF1, and ZRSR2 and Response to Decitabine in Myelodysplastic Syndrome. , 2015, Anticancer research.

[27]  M. Treppendahl,et al.  Hypomethylation and up-regulation of PD-1 in T cells by azacytidine in MDS/AML patients: A rationale for combined targeting of PD-1 and DNA methylation , 2015, Oncotarget.

[28]  M. Lübbert,et al.  Combining DNA methyltransferase and histone deacetylase inhibition to treat acute myeloid leukemia/myelodysplastic syndrome: Achievements and challenges , 2015, Cancer.

[29]  E. Solary,et al.  Chronic myelomonocytic leukemia in younger patients: molecular and cytogenetic predictors of survival and treatment outcome , 2015, Blood Cancer Journal.

[30]  A. Ganser,et al.  Decitabine improves progression-free survival in older high-risk MDS patients with multiple autosomal monosomies: results of a subgroup analysis of the randomized phase III study 06011 of the EORTC Leukemia Cooperative Group and German MDS Study Group , 2015, Annals of Hematology.

[31]  R. Bejar,et al.  Recent developments in myelodysplastic syndromes. , 2014, Blood.

[32]  D. Neuberg,et al.  TET2 mutations predict response to hypomethylating agents in myelodysplastic syndrome patients. , 2014, Blood.

[33]  T. Haferlach,et al.  Response to azacitidine is independent of p53 expression in higher-risk myelodysplastic syndromes and secondary acute myeloid leukemia , 2014, Haematologica.

[34]  D. Neuberg,et al.  Somatic mutations predict poor outcome in patients with myelodysplastic syndrome after hematopoietic stem-cell transplantation. , 2014, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[35]  C. Preudhomme,et al.  Prognostic value of TP53 gene mutations in myelodysplastic syndromes and acute myeloid leukemia treated with azacitidine. , 2014, Leukemia research.

[36]  P. Rohrlich,et al.  Phenotypic and genotypic characterization of azacitidine-sensitive and resistant SKM1 myeloid cell lines , 2014, Oncotarget.

[37]  P. Fenaux,et al.  Epigenetics of myelodysplastic syndromes , 2014, Leukemia.

[38]  M. Treppendahl,et al.  5-Azacytidine treatment sensitizes tumor cells to T-cell mediated cytotoxicity and modulates NK cells in patients with myeloid malignancies , 2014, Blood Cancer Journal.

[39]  M. Konopleva,et al.  Lack of association of IDH1, IDH2 and DNMT3A mutations with outcome in older patients with acute myeloid leukemia treated with hypomethylating agents , 2014, Leukemia & lymphoma.

[40]  M. Lübbert,et al.  Clinical development of demethylating agents in hematology. , 2014, The Journal of clinical investigation.

[41]  S. Parmar,et al.  Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents , 2013, Leukemia.

[42]  C Haferlach,et al.  Landscape of genetic lesions in 944 patients with myelodysplastic syndromes , 2013, Leukemia.

[43]  A. Jankowska,et al.  Impact of molecular mutations on treatment response to DNMT inhibitors in myelodysplasia and related neoplasms , 2014, Leukemia.

[44]  M. Stratton,et al.  Clinical and biological implications of driver mutations in myelodysplastic syndromes. , 2013, Blood.

[45]  G. Mufti,et al.  Comprehensive Mutational Screening Of 5-Azacitidne Treated Myelodysplastic Syndrome (MDS) Patients Fails To Identify a Specific Mutational Profile Predicting Response To Therapy , 2013 .

[46]  G. Mufti,et al.  Lenalidomide as a disease-modifying agent in patients with del(5q) myelodysplastic syndromes: linking mechanism of action to clinical outcomes , 2013, Annals of Hematology.

[47]  Dean Anthony Lee,et al.  Decitabine has a biphasic effect on natural killer cell viability, phenotype, and function under proliferative conditions. , 2013, Molecular immunology.

[48]  Y. Saunthararajah,et al.  Gender, Cytidine Deaminase, and 5-Aza/Decitabine—Response , 2013, Clinical Cancer Research.

[49]  Y. Tohyama,et al.  Effects of DNA methyltransferase inhibitors (DNMTIs) on MDS-derived cell lines. , 2013, Experimental hematology.

[50]  M. Maio,et al.  Immunomodulatory activity of SGI-110, a 5-aza-2′-deoxycytidine-containing demethylating dinucleotide , 2013, Cancer Immunology, Immunotherapy.

[51]  Luca Malcovati,et al.  Revised international prognostic scoring system for myelodysplastic syndromes. , 2012, Blood.

[52]  M. Minden,et al.  Multicenter, randomized, open-label, phase III trial of decitabine versus patient choice, with physician advice, of either supportive care or low-dose cytarabine for the treatment of older patients with newly diagnosed acute myeloid leukemia. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[53]  C. Bloomfield,et al.  DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia , 2012, Leukemia.

[54]  C. Steidl,et al.  New comprehensive cytogenetic scoring system for primary myelodysplastic syndromes (MDS) and oligoblastic acute myeloid leukemia after MDS derived from an international database merge. , 2012, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[55]  M. Mohty,et al.  Impact of the hypomethylating agent 5-azacytidine on dendritic cells function. , 2011, Experimental hematology.

[56]  M. Cazzola,et al.  A randomized phase 3 study of lenalidomide versus placebo in RBC transfusion-dependent patients with Low-/Intermediate-1-risk myelodysplastic syndromes with del5q. , 2011, Blood.

[57]  A. Vekhoff,et al.  Molecular predictors of response to decitabine in advanced chronic myelomonocytic leukemia: a phase 2 trial. , 2011, Blood.

[58]  M. Vignetti,et al.  Role of BCL2L10 methylation and TET2 mutations in higher risk myelodysplastic syndromes treated with 5-Azacytidine , 2011, Leukemia.

[59]  D. Neuberg,et al.  Clinical effect of point mutations in myelodysplastic syndromes. , 2011, The New England journal of medicine.

[60]  B. Quesnel,et al.  Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias , 2011, Leukemia.

[61]  U. Germing,et al.  Treatment of poor-risk myelodysplastic syndromes and acute myeloid leukemia with a combination of 5-azacytidine and valproic acid , 2011, Clinical Epigenetics.

[62]  F. Lyko,et al.  Azacytidine and Decitabine Induce Gene-Specific and Non-Random DNA Demethylation in Human Cancer Cell Lines , 2011, PloS one.

[63]  A. Stamatoullas,et al.  Prognostic factors for response and overall survival in 282 patients with higher-risk myelodysplastic syndromes treated with azacitidine. , 2011, Blood.

[64]  R. Arceci Impact of TET2 mutations on response rate to azacitidine in myelodysplastic syndromes and low blast count acute myeloid leukemias , 2011 .

[65]  J. Karbach,et al.  The DNA demethylating agent 5-aza-2'-deoxycytidine induces expression of NY-ESO-1 and other cancer/testis antigens in myeloid leukemia cells. , 2010, Leukemia research.

[66]  Helen Brady,et al.  A Comparison of Azacitidine and Decitabine Activities in Acute Myeloid Leukemia Cell Lines , 2010, PloS one.

[67]  H. Dombret,et al.  Azacitidine prolongs overall survival compared with conventional care regimens in elderly patients with low bone marrow blast count acute myeloid leukemia. , 2010, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[68]  A. Hagemeijer,et al.  Influence of Karyotype On Overall Survival in Patients with Higher-Risk Myelodysplastic Syndrome Treated with Azacitidine or a Conventional Care Regimen. , 2009 .

[69]  Valeria Santini,et al.  Efficacy of azacitidine compared with that of conventional care regimens in the treatment of higher-risk myelodysplastic syndromes: a randomised, open-label, phase III study. , 2009, The Lancet. Oncology.

[70]  C. Plass,et al.  The DNA methyltransferase inhibitors azacitidine, decitabine and zebularine exert differential effects on cancer gene expression in acute myeloid leukemia cells , 2009, Leukemia.

[71]  G. Mufti,et al.  CDKN2B methylation status and isolated chromosome 7 abnormalities predict responses to treatment with 5-azacytidine , 2007, Leukemia.

[72]  R. Larson,et al.  Further analysis of trials with azacitidine in patients with myelodysplastic syndrome: studies 8421, 8921, and 9221 by the Cancer and Leukemia Group B. , 2006, Journal of clinical oncology : official journal of the American Society of Clinical Oncology.

[73]  B. Cheson,et al.  Clinical application and proposal for modification of the International Working Group (IWG) response criteria in myelodysplasia. , 2006, Blood.

[74]  John M Bennett,et al.  Decitabine improves patient outcomes in myelodysplastic syndromes , 2006, Cancer.

[75]  W. Hiddemann,et al.  Risk assessment by monitoring expression levels of partial tandem duplications in the MLL gene in acute myeloid leukemia during therapy. , 2005, Haematologica.

[76]  Martin Dugas,et al.  Analysis of FLT3 length mutations in 1003 patients with acute myeloid leukemia: correlation to cytogenetics, FAB subtype, and prognosis in the AMLCG study and usefulness as a marker for the detection of minimal residual disease. , 2002 .

[77]  M. Lübbert,et al.  Cytogenetic responses in high‐risk myelodysplastic syndrome following low‐dose treatment with the DNA methylation inhibitor 5‐aza‐2′‐deoxycytidine , 2001, British journal of haematology.