Preleukemic mutations in human acute myeloid leukemia affect epigenetic regulators and persist in remission

Significance A growing body of evidence has determined that somatic mutations in acute myeloid leukemia (AML) accumulate in self-renewing hematopoietic stem cells (HSCs). Thus, at the time of diagnosis, AML patients harbor preleukemic HSCs containing some, but not all, of the mutations in the downstream leukemia. Despite these findings, common patterns of preleukemic clonal evolution have not been determined, nor has the response of preleukemic HSCs to standard therapy been identified. This report addresses both of these questions determining that there are common patterns of preleukemic clonal evolution in AML, and that these preleukemic HSCs often survive standard induction chemotherapy. This study is of interest to the AML field, and broadly in cancer genomics as the principle that stem cells acquire initial cancer-initiating mutations is likely to extend beyond AML. Cancer is widely characterized by the sequential acquisition of genetic lesions in a single lineage of cells. Our previous studies have shown that, in acute myeloid leukemia (AML), mutation acquisition occurs in functionally normal hematopoietic stem cells (HSCs). These preleukemic HSCs harbor some, but not all, of the mutations found in the leukemic cells. We report here the identification of patterns of mutation acquisition in human AML. Our findings support a model in which mutations in “landscaping” genes, involved in global chromatin changes such as DNA methylation, histone modification, and chromatin looping, occur early in the evolution of AML, whereas mutations in “proliferative” genes occur late. Additionally, we analyze the persistence of preleukemic mutations in patients in remission and find CD34+ progenitor cells and various mature cells that harbor preleukemic mutations. These findings indicate that preleukemic HSCs can survive induction chemotherapy, identifying these cells as a reservoir for the reevolution of relapsed disease. Finally, through the study of several cases of relapsed AML, we demonstrate various evolutionary patterns for the generation of relapsed disease and show that some of these patterns are consistent with involvement of preleukemic HSCs. These findings provide key insights into the monitoring of minimal residual disease and the identification of therapeutic targets in human AML.

[1]  Asim Khwaja,et al.  Acute myeloid leukaemia , 2016, Nature Reviews Disease Primers.

[2]  Lloyd H. Michael,et al.  The Guide for the Care and Use of Laboratory Animals. , 2016, ILAR journal.

[3]  Uku. Innere Medizin Clonal evolution in relapsed NPM1 mutated acute myeloid leukemia , 2014 .

[4]  David T. W. Jones,et al.  Signatures of mutational processes in human cancer , 2013, Nature.

[5]  Benjamin J. Raphael,et al.  Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. , 2013, The New England journal of medicine.

[6]  R. Majeti,et al.  Clonal evolution of acute leukemia genomes , 2013, Oncogene.

[7]  Mithat Gonen,et al.  Recurrent Somatic TET2 Mutations in Normal Elderly Individuals With Clonal Hematopoiesis , 2012, Nature Genetics.

[8]  I. Weissman,et al.  Clonal Evolution of Preleukemic Hematopoietic Stem Cells Precedes Human Acute Myeloid Leukemia , 2012, Science Translational Medicine.

[9]  Joshua F. McMichael,et al.  The Origin and Evolution of Mutations in Acute Myeloid Leukemia , 2012, Cell.

[10]  Joshua F. McMichael,et al.  Clonal evolution in relapsed acute myeloid leukemia revealed by whole genome sequencing , 2011, Nature.

[11]  J. Licht,et al.  DNMT3A mutations in acute myeloid leukemia , 2011, Nature Genetics.

[12]  Yong-mei Zhu,et al.  Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia , 2011, Nature Genetics.

[13]  Ash A. Alizadeh,et al.  Prospective separation of normal and leukemic stem cells based on differential expression of TIM3, a human acute myeloid leukemia stem cell marker , 2011, Proceedings of the National Academy of Sciences.

[14]  H. Drexler,et al.  Inactivating mutations of the histone methyltransferase gene EZH2 in myeloid disorders , 2010, Nature Genetics.

[15]  R. Kuiper,et al.  Somatic mutations of the histone methyltransferase gene EZH2 in myelodysplastic syndromes , 2010, Nature Genetics.

[16]  I. Tikhonova,et al.  Genetic diagnosis by whole exome capture and massively parallel DNA sequencing , 2009, Proceedings of the National Academy of Sciences.

[17]  Ken Chen,et al.  Recurring mutations found by sequencing an acute myeloid leukemia genome. , 2009, The New England journal of medicine.

[18]  D. Gilliland,et al.  Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. , 2009, Blood.

[19]  I. Weissman,et al.  Glycogen synthase kinase 3β missplicing contributes to leukemia stem cell generation , 2009, Proceedings of the National Academy of Sciences.

[20]  Emily H Turner,et al.  Targeted Capture and Massively Parallel Sequencing of Twelve Human Exomes , 2009, Nature.

[21]  Amy E. Hawkins,et al.  DNA sequencing of a cytogenetically normal acute myeloid leukemia genome , 2008, Nature.

[22]  I. Weissman,et al.  Identification of a hierarchy of multipotent hematopoietic progenitors in human cord blood. , 2007, Cell stem cell.

[23]  I. Weissman,et al.  Stem cell research: paths to cancer therapies and regenerative medicine. , 2005, JAMA.

[24]  Lucio Luzzatto,et al.  A quantitative measurement of the human somatic mutation rate. , 2005, Cancer research.

[25]  Laurie E Ailles,et al.  Granulocyte-macrophage progenitors as candidate leukemic stem cells in blast-crisis CML. , 2004, The New England journal of medicine.

[26]  H. Kantarjian,et al.  Acute myeloid leukemia , 2018, Methods in Molecular Biology.

[27]  I. Weissman,et al.  AML1/ETO-expressing nonleukemic stem cells in acute myelogenous leukemia with 8;21 chromosomal translocation. , 2000, Proceedings of the National Academy of Sciences of the United States of America.