Skip to main content Accessibility help
×
Hostname: page-component-8448b6f56d-42gr6 Total loading time: 0 Render date: 2024-04-24T19:45:09.735Z Has data issue: false hasContentIssue false

69 - The molecular basis of acute myeloid leukemia

from Part 3.6 - Molecular pathology: lymphoma and leukemia

Published online by Cambridge University Press:  05 February 2015

Kim L. Rice
Affiliation:
Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
Monica Buzzai
Affiliation:
Novartis, Origgio, VA, Italy
Jessica Altman
Affiliation:
Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
Jonathan D. Licht
Affiliation:
Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
Edward P. Gelmann
Affiliation:
Columbia University, New York
Charles L. Sawyers
Affiliation:
Memorial Sloan-Kettering Cancer Center, New York
Frank J. Rauscher, III
Affiliation:
The Wistar Institute Cancer Centre, Philadelphia
Get access

Summary

Introduction

Acute myeloid leukemia (AML) is an oligoclonal malignancy characterized by several differences from normal hematopoiesis, including blockade of normal differentiation, enhanced self-renewal, increased proliferation, impaired apoptosis, dissemination, and genomic instability. Recent advances in our understanding of the pathogenesis of AML have involved elucidation of the role of aberrant chromosomal rearrangement, amplifications, deletions, and point mutations, and aberrant regulation of gene expression, governed in part by changes in chromatin. AML can now be subclassified based upon gene mutations, gene expression, miRNA expression, and DNA methylation states, and many of the phenotypic properties of AML and alterations in gene expression can be mapped onto underlying genetic lesions. Within several years, a near complete categorization of AML will be achieved, and a variety of new therapeutic targets will be identified. Remaining challenges will be to understand the molecular mechanisms linking genetic and epigenetic changes to leukemia cell growth and the translation of these findings into specific therapies that may target mutant proteins or deregulated gene networks.

The evolving modes of classification of acute myeloid leukemia

The French-American-British (FAB) system classified acute myeloid leukemia (AML) by analogy to normal myelopoiesis, but yielded relatively poor information in that only a few subtypes, such as acute promyelocytic leukemia (APL), could be distinguished as having a distinct prognosis. Over the past two decades, conventional cytogenetics, flow cytometry, fluorescence in situ hybridization, DNA sequencing, and PCR have led to the definition of more precise subsets of AML with distinct prognoses. This was reflected in the 1999 WHO system that includes cytogenetic and molecular anomalies (1). The revolution in genomic technology will soon lead to a reassessment of AML and the definition of even smaller prognostic subsets.

Type
Chapter
Information
Molecular Oncology
Causes of Cancer and Targets for Treatment
, pp. 751 - 768
Publisher: Cambridge University Press
Print publication year: 2013

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

References

Vardiman, JW, Harris, NL, Brunning, RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood 2002;100:2292–302.CrossRef
Grimwade, D, Walker, H, Oliver, F, et al. The importance of diagnostic cytogenetics on outcome in AML: analysis of 1,612 patients entered into the MRC AML 10 trial. The Medical Research Council Adult and Children's Leukaemia Working Parties. Blood 1998;92:2322–33.
Marcucci, G, Haferlach, T, Dohner, H.Molecular genetics of adult acute myeloid leukemia: prognostic and therapeutic implications. Journal of Clinical Oncology 2011;29:475–86.CrossRefGoogle ScholarPubMed
Paschka, P, Marcucci, G, Ruppert, AS, et al. Adverse prognostic significance of KIT mutations in adult acute myeloid leukemia with inv(16) and t(8;21): a Cancer and Leukemia Group B Study. Journal of Clinical Oncology 2006;24:3904–11.CrossRefGoogle Scholar
Gale, RE, Green, C, Allen, C, et al. The impact of FLT3 internal tandem duplication mutant level, number, size, and interaction with NPM1 mutations in a large cohort of young adult patients with acute myeloid leukemia. Blood 2008;111:2776–84.CrossRef
Meyer, C, Kowarz, E, Hofmann, J, et al. New insights to the MLL recombinome of acute leukemias. Leukemia 2009;23:1490–9.CrossRef
Caligiuri, MA, Schichman, SA, Strout, MP, et al. Molecular rearrangement of the ALL-1 gene in acute myeloid leukemia without cytogenetic evidence of 11q23 chromosomal translocations. Cancer Research 1994;54:370–3.
Whitman, SP, Hackanson, B, Liyanarachchi, S, et al. DNA hypermethylation and epigenetic silencing of the tumor suppressor gene, SLC5A8, in acute myeloid leukemia with the MLL partial tandem duplication. Blood 2008;112:2013–16.CrossRef
Vardiman, JW, Thiele, J, Arber, DA, et al. The 2008 revision of the World Health Organization (WHO) classification of myeloid neoplasms and acute leukemia: rationale and important changes. Blood 2009;114:937–51.CrossRef
Renneville, A, Boissel, N, Zurawski, V, et al. Wilms tumor 1 gene mutations are associated with a higher risk of recurrence in young adults with acute myeloid leukemia: a study from the Acute Leukemia French Association. Cancer 2009;115:3719–27.CrossRef
Schnittger, S, Dicker, F, Kern, W, et al. RUNX1 mutations are frequent in de novo AML with non complex karyotype and confer an unfavourable prognosis. Blood 2011;117:2348–57.CrossRef
Falini, B, Mecucci, C, Tiacci, E, et al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. New England Journal of Medicine 2005;352:254–66.CrossRefGoogle ScholarPubMed
Frohling, S, Schlenk, RF, Stolze, I, et al. CEBPA mutations in younger adults with acute myeloid leukemia and normal cytogenetics: prognostic relevance and analysis of cooperating mutations. Journal of Clinical Oncology 2004;22:624–33.CrossRefGoogle ScholarPubMed
Lin, LI, Chen, CY, Lin, DT, et al. Characterization of CEBPA mutations in acute myeloid leukemia: most patients with CEBPA mutations have biallelic mutations and show a distinct immunophenotype of the leukemic cells. Clinical Cancer Research 2005;11:1372–9.CrossRef
Wouters, BJ, Lowenberg, B, Erpelinck-Verschueren, CA, et al. Double CEBPA mutations, but not single CEBPA mutations, define a subgroup of acute myeloid leukemia with a distinctive gene expression profile that is uniquely associated with a favorable outcome. Blood 2009;113:3088–91.CrossRef
Metzeler, KH, A, Benthaus, T, et al. ERG expression is an independent prognostic factor and allows refined risk stratification in cytogenetically normal acute myeloid leukemia: a comprehensive analysis of ERG, MN1, and BAALC transcript levels using oligonucleotide microarrays. Journal of Clinical Oncology 2009;27:5031–8.CrossRefGoogle ScholarPubMed
Groschel, S, Lugthart, S, Schlenk, RF, et al. High EVI1 expression predicts outcome in younger adult patients with acute myeloid leukemia and is associated with distinct cytogenetic abnormalities. Journal of Clinical Oncology 2010;28:2101–7.CrossRefGoogle ScholarPubMed
Grimwade, D, Hills, RK, Moorman, AV, et al. Refinement of cytogenetic classification in acute myeloid leukemia: determination of prognostic significance of rare recurring chromosomal abnormalities among 5876 younger adult patients treated in the United Kingdom Medical Research Council trials. Blood 2010;116:354–65.CrossRef
Haferlach, C, Dicker, F, Herholz, H, et al. Mutations of the TP53 gene in acute myeloid leukemia are strongly associated with a complex aberrant karyotype. Leukemia 2008;22:1539–41.CrossRef
Breems, DA, Van Putten, WL, De Greef, GE, et al. Monosomal karyotype in acute myeloid leukemia: a better indicator of poor prognosis than a complex karyotype. Journal of Clinical Oncology 2008;26:4791–7.CrossRefGoogle ScholarPubMed
Medeiros, BC, Othus, M, Fang, M, Roulston, D, Appelbaum FR. Prognostic impact of monosomal karyotype in young adult and elderly acute myeloid leukemia: the Southwest Oncology Group (SWOG) experience. Blood 2010;116:2224–8.CrossRef
Raghavan, M, Lillington, DM, Skoulakis, S, et al. Genome-wide single nucleotide polymorphism analysis reveals frequent partial uniparental disomy due to somatic recombination in acute myeloid leukemias. Cancer Research 2005;65:375–8.
Suela, J, Alvarez, S, Cifuentes, F, et al. DNA profiling analysis of 100 consecutive de novo acute myeloid leukemia cases reveals patterns of genomic instability that affect all cytogenetic risk groups. Leukemia 2007;21:1224–31.CrossRef
Bullinger, L, Kronke, J, Schon, C, et al. Identification of acquired copy number alterations and uniparental disomies in cytogenetically normal acute myeloid leukemia using high-resolution single-nucleotide polymorphism analysis. Leukemia 2010;24:438–49.CrossRef
Delhommeau, F, Dupont, S, Della Valle, V, et al. Mutation in TET2 in myeloid cancers. New England Journal of Medicine 2009;360:2289–301.CrossRefGoogle ScholarPubMed
Chou, WC, Chou, SC, Liu, CY, et al. TET2 mutation is an unfavorable prognostic factor in acute myeloid leukemia patients with intermediate-risk cytogenetics. Blood 2011;118:3803–10.CrossRef
Ley, TJ, Ding, L, Walter, MJ, et al. DNMT3A mutations in acute myeloid leukemia. New England Journal of Medicine 2010;363:2424–33.CrossRefGoogle ScholarPubMed
Thol, F, Damm, F, Ludeking, A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. Journal of Clinical Oncology 2011;29:2889–96.CrossRefGoogle ScholarPubMed
Mardis, ER, Ding, L, Dooling, DJ, et al. Recurring mutations found by sequencing an acute myeloid leukemia genome. New England Journal of Medicine 2009;361:1058–66.CrossRefGoogle ScholarPubMed
Marcucci, G, Maharry, K, Wu, YZ, et al. IDH1 and IDH2 gene mutations identify novel molecular subsets within de novo cytogenetically normal acute myeloid leukemia: a Cancer and Leukemia Group B study. Journal of Clinical Oncology 2010;28:2348–55.CrossRefGoogle ScholarPubMed
Boissel, N, Nibourel, O, Renneville, A, et al. Prognostic impact of isocitrate dehydrogenase enzyme isoforms 1 and 2 mutations in acute myeloid leukemia: a study by the Acute Leukemia French Association group. Journal of Clinical Oncology 2010;28:3717–23.CrossRefGoogle ScholarPubMed
Carbuccia, N, Trouplin, V, Gelsi-Boyer, V, et al. Mutual exclusion of ASXL1 and NPM1 mutations in a series of acute myeloid leukemias. Leukemia 2010;24:469–73.CrossRef
Yoshida, K, Sanada, M, Shiraishi, Y, et al. Frequent pathway mutations of splicing machinery in myelodysplasia. Nature 2011;478:64–9.CrossRef
Haferlach T, Kohlmann A, Schnittger S, et al. AML M3 and AML M3 variant each have a distinct gene expression signature but also share patterns different from other genetically defined AML subtypes. Genes, Chromosomes and Cancer 2005;43:113–27.CrossRef
Ebert, BL, Golub, TR. Genomic approaches to hematologic malignancies. Blood 2004;104:923–32.CrossRef
Valk, PJ, Verhaak, RG, Beijen, MA, et al. Prognostically useful gene-expression profiles in acute myeloid leukemia. New England Journal of Medicine 2004;350:1617–28.CrossRefGoogle ScholarPubMed
Bullinger, L, Dohner, K, Bair, E, et al. Use of gene-expression profiling to identify prognostic subclasses in adult acute myeloid leukemia. New England Journal of Medicine 2004;350:1605–16.CrossRefGoogle ScholarPubMed
Figueroa, ME, Reimers, M, Thompson, RF, et al. An integrative genomic and epigenomic approach for the study of transcriptional regulation. PLoS ONE 2008;3:e1882.
Figueroa, ME, Lugthart, S, Li, Y, et al. DNA methylation signatures identify biologically distinct subtypes in acute myeloid leukemia. Cancer Cell 2010;17:13–27.CrossRef
Muller-Tidow, C, Klein, HU, Hascher, A, et al. Profiling of histone H3 lysine 9 trimethylation levels predicts transcription factor activity and survival in acute myeloid leukemia. Blood 2010;116:3564–71.CrossRef
Lu, J, Getz, G, Miska, EA, et al. MicroRNA expression profiles classify human cancers. Nature 2005;435:834–8.CrossRef
Li, Z, Lu, J, Sun, M, et al. Distinct microRNA expression profiles in acute myeloid leukemia with common translocations. Proceedings of the National Academy of Sciences USA 2008;105:15 535–40.
Garzon, R, Volinia, S, Liu, CG, et al. MicroRNA signatures associated with cytogenetics and prognosis in acute myeloid leukemia. Blood 2008;111:3183–9.CrossRef
Marcucci, G, Radmacher, MD, Maharry, K, et al. MicroRNA expression in cytogenetically normal acute myeloid leukemia. New England Journal of Medicine 2008;358:1919–28.CrossRefGoogle ScholarPubMed
Fernandez, HF, Sun, Z, Yao, X, et al. Anthracycline dose intensification in acute myeloid leukemia. New England Journal of Medicine 2009;361:1249–59.CrossRefGoogle ScholarPubMed
Schlenk, RF, Dohner, K, Krauter, J, et al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. New England Journal of Medicine 2008;358:1909–18.CrossRefGoogle ScholarPubMed
Green, CL, Koo, KK, Hills, RK, et al. Prognostic significance of CEBPA mutations in a large cohort of younger adult patients with acute myeloid leukemia: impact of double CEBPA mutations and the interaction with FLT3 and NPM1 mutations. Journal of Clinical Oncology 2010;28:2739–47.CrossRefGoogle Scholar
Marková, J, Michková, P, Burčková, K, et al. Prognostic impact of DNMT3A mutations in patients with intermediate cytogenetic risk profile acute myeloid leukemia. European Journal of Haematology 2011;88:128–35.CrossRefGoogle ScholarPubMed
Lapidot, T, Sirard, C, Vormoor, J, et al. A cell initiating human acute myeloid leukaemia after transplantation into SCID mice. Nature 1994;367:645–8.CrossRef
Guan, Y, Gerhard, B, Hogge, DE. Detection, isolation, and stimulation of quiescent primitive leukemic progenitor cells from patients with acute myeloid leukemia (AML). Blood 2003;101:3142–9.CrossRef
Terpstra, W, Ploemacher, RE, Prins, A, et al. Fluorouracil selectively spares acute myeloid leukemia cells with long-term growth abilities in immunodeficient mice and in culture. Blood 1996;88:1944–50.
McDermott, SP, Eppert, K, Lechman, ER, Doedens, M, Dick, JE. Comparison of human cord blood engraftment between immunocompromised mouse strains. Blood 2010;116:193–200.CrossRef
Kelly, PN, Dakic, A, Adams, JM, Nutt, SL, Strasser, A. Tumor growth need not be driven by rare cancer stem cells. Science 2007;317:337.CrossRef
Eppert, K, Takenaka, K, Lechman, ER, et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nature Medicine 2011;17:1086–93.CrossRef
Quintana, E, Shackleton, M, Sabel, MS, et al. Efficient tumour formation by single human melanoma cells. Nature 2008;456:593–8.CrossRef
Kottaridis, PD, Gale, RE, Langabeer, SE, et al. Studies of FLT3 mutations in paired presentation and relapse samples from patients with acute myeloid leukemia: implications for the role of FLT3 mutations in leukemogenesis, minimal residual disease detection, and possible therapy with FLT3 inhibitors. Blood 2002;100:2393–8.CrossRef
Barabe, F, Kennedy, JA, Hope, KJ, Dick, JE. Modeling the initiation and progression of human acute leukemia in mice. Science 2007;316:600–4.CrossRef
Anderson, K, Lutz, C, van Delft, FW, et al. Genetic variegation of clonal architecture and propagating cells in leukaemia. Nature 2011;469:356–61.CrossRef
Alcalay, M, Tiacci, E, Bergomas, R, et al. Acute myeloid leukemia bearing cytoplasmic nucleophosmin (NPMc+ AML) shows a distinct gene expression profile characterized by up-regulation of genes involved in stem-cell maintenance. Blood 2005;106:899–902.CrossRef
Alcalay, M, Meani, N, Gelmetti, V, et al. Acute myeloid leukemia fusion proteins deregulate genes involved in stem cell maintenance and DNA repair. Journal of Clinical Investigation 2003;112:1751–61.
Chung, KY, Morrone, G, Schuringa, JJ, et al. Enforced expression of an Flt3 internal tandem duplication in human CD34+ cells confers properties of self-renewal and enhanced erythropoiesis. Blood 2005;105:77–84.CrossRef
Muller-Tidow, C, Steffen, B, Cauvet, T, et al. Translocation products in acute myeloid leukemia activate the Wnt signaling pathway in hematopoietic cells. Molecular and Cellular Biology 2004;24:2890–904.CrossRef
Reya, T, Duncan, AW, Ailles, L, et al. A role for Wnt signalling in self-renewal of haematopoietic stem cells. Nature 2003;423:409–14.CrossRef
Gentles, AJ, Plevritis, SK, Majeti, R, Alizadeh, AA.Association of a leukemic stem cell gene expression signature with clinical outcomes in acute myeloid leukemia. Journal of the American Medical Association 2010;304:2706–15.CrossRefGoogle ScholarPubMed
Nakao, M, Yokota, S, Iwai, T, et al. Internal tandem duplication of the flt3 gene found in acute myeloid leukemia. Leukemia 1996;10:1911–18.
Breitenbuecher, F, Schnittger, S, Grundler, R, et al. Identification of a novel type of ITD mutations located in nonjuxtamembrane domains of the FLT3 tyrosine kinase receptor. Blood 2009;113:4074–7.CrossRef
Yamamoto, Y, Kiyoi, H, Nakano, Y, et al. Activating mutation of D835 within the activation loop of FLT3 in human hematologic malignancies. Blood 2001;97:2434–9.CrossRef
Lee, BH, Tothova, Z, Levine, RL, et al. FLT3 mutations confer enhanced proliferation and survival properties to multipotent progenitors in a murine model of chronic myelomonocytic leukemia. Cancer Cell 2007;12:367–80.CrossRef
Cammenga, J, Horn, S, Bergholz, U, et al. Extracellular KIT receptor mutants, commonly found in core binding factor AML, are constitutively active and respond to imatinib mesylate. Blood 2005;106:3958–61.CrossRef
Gari, M, Goodeve, A, Wilson, G, et al. c-kit proto-oncogene exon 8 in-frame deletion plus insertion mutations in acute myeloid leukaemia. British Journal of Haematology 1999;105:894–900.CrossRefGoogle ScholarPubMed
Luck, SC, Russ, AC, Du, J, et al. KIT mutations confer a distinct gene expression signature in core binding factor leukaemia. British Journal of Haematology 2010;148:925–37.CrossRefGoogle Scholar
Steensma, DP, Dewald, GW, Lasho, TL, et al. The JAK2 V617F activating tyrosine kinase mutation is an infrequent event in both “atypical” myeloproliferative disorders and myelodysplastic syndromes. Blood 2005;106:1207–9.CrossRef
Steensma, DP, McClure, RF, Karp, JE, et al. JAK2 V617F is a rare finding in de novo acute myeloid leukemia, but STAT3 activation is common and remains unexplained. Leukemia 2006;20:971–8.CrossRef
Dohner, K, Du, J, Corbacioglu, A, et al. JAK2V617F mutations as cooperative genetic lesions in t(8;21)-positive acute myeloid leukemia. Haematologica 2006;91:1569–70.
Tomasson, MH, Xiang, Z, Walgren, R, et al. Somatic mutations and germline sequence variants in the expressed tyrosine kinase genes of patients with de novo acute myeloid leukemia. Blood 2008;111:4797–808.CrossRef
Bowen, DT, Frew, ME, Hills, R, et al. RAS mutation in acute myeloid leukemia is associated with distinct cytogenetic subgroups but does not influence outcome in patients younger than 60 years. Blood 2005;106:2113–19.CrossRef
Shih, LY, Huang, CF, Wang, PN, et al. Acquisition of FLT3 or N-ras mutations is frequently associated with progression of myelodysplastic syndrome to acute myeloid leukemia. Leukemia 2004;18:466–75.CrossRef
Sargin, B, Choudhary, C, Crosetto, N, et al. Flt3-dependent transformation by inactivating c-Cbl mutations in AML. Blood 2007;110:1004–12.CrossRef
Mulloy, JC, Jankovic, V, Wunderlich, M, et al. AML1-ETO fusion protein up-regulates TRKA mRNA expression in human CD34+ cells, allowing nerve growth factor-induced expansion. Proceedings of the National Academy of Sciences USA 2005;102:4016–21.CrossRef
Kentsis, A, Sanda, T, Reed, C, et al. Combined targeting of the MET and FGF receptor tyrosine kinases induces sustained AML cell death by preventing compensatory upregulation of HGF in response to MET kinase inhibition. ASH Annual Meeting Abstracts 2011;118:1405.
Mi, S, Lu, J, Sun, M, et al. MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia. Proceedings of the National Academy of Sciences USA 2007;104:19 971–6.
Bai, H, Xu, R, Cao, Z, Wei, D, Wang, C. Involvement of miR-21 in resistance to daunorubicin by regulating PTEN expression in the leukaemia K562 cell line. FEBS Letters 2011;585:402–8.CrossRef
Fazi, F, Zardo, G, Gelmetti, V, et al. Heterochromatic gene repression of the retinoic acid pathway in acute myeloid leukemia. Blood 2007;109:4432–40.CrossRef
Osato, M. Point mutations in the RUNX1/AML1 gene: another actor in RUNX leukemia. Oncogene 2004;23:4284–96.CrossRef
Cook, WD, McCaw BJ, Herring C, et al. PU.1 is a suppressor of myeloid leukemia, inactivated in mice by gene deletion and mutation of its DNA binding domain. Blood 2004;104:3437–44.CrossRef
Melnick, A, Licht, JD. Deconstructing a disease: RARalpha, its fusion partners, and their roles in the pathogenesis of acute promyelocytic leukemia. Blood 1999;93:3167–215.
Zhu, J, Nasr, R, Peres, L, et al. RXR is an essential component of the oncogenic PML/RARA complex in vivo. Cancer Cell 2007;12:23–35.CrossRef
Zhou, J, Peres, L, Honore, N, et al. Dimerization-induced corepressor binding and relaxed DNA-binding specificity are critical for PML/RARA-induced immortalization. Proceedings of the National Academy of Sciences USA 2006;103:9238–43.CrossRef
Park, DJ, Chumakov, AM, Vuong, PT, et al. CCAAT/enhancer binding protein epsilon is a potential retinoid target gene in acute promyelocytic leukemia treatment. Journal of Clinical Investigation 1999;103:1399–408.CrossRefGoogle ScholarPubMed
Zheng, X, Beissert, T, Kukoc-Zivojnov, N, et al. Gamma-catenin contributes to leukemogenesis induced by AML-associated translocation products by increasing the self-renewal of very primitive progenitor cells. Blood 2004;103:3535–43.CrossRef
Lallemand-Breitenbach, V, de The, H. PML nuclear bodies. Cold Spring Harbor Perspectives in Biology 2010;2:a000661.
Borden, KL. Pondering the puzzle of PML (promyelocytic leukemia) nuclear bodies: can we fit the pieces together using an RNA regulon? Biochimica et Biophysica Acta 2008;1783:2145–54.
Xu, ZX, Timanova-Atanasova, A, Zhao, RX, Chang, KS. PML colocalizes with and stabilizes the DNA damage response protein TopBP1. Molecular and Cellular Biology 2003;23:4247–56.CrossRef
Degos, L, Dombret, H, Chomienne, C, et al. All-trans-retinoic acid as a differentiating agent in the treatment of acute promyelocytic leukemia. Blood 1995;85:2643–53.
Tallman, MS. Treatment of relapsed or refractory acute promyelocytic leukemia. Best Practice and Research in Clinical Haematology 2007;20:57–65.CrossRef
Hu, J, Liu, YF, Wu, CF, et al. Long-term efficacy and safety of all-trans retinoic acid/arsenic trioxide-based therapy in newly diagnosed acute promyelocytic leukemia. Proceedings of the National Academy of Sciences USA 2009;106:3342–7.CrossRef
Jeanne, M, Lallemand-Breitenbach, V, Ferhi, O, et al. PML/RARA oxidation and arsenic binding initiate the antileukemia response of As2O3. Cancer Cell 2010;18:88–98.CrossRef
Zhang, XW, Yan, XJ, Zhou, ZR, et al. Arsenic trioxide controls the fate of the PML-RARalpha oncoprotein by directly binding PML. Science 2010;328:240–3.CrossRef
de The, H, Chen, Z. Acute promyelocytic leukaemia: novel insights into the mechanisms of cure. Nature Reviews Cancer 2010;10:775–83.CrossRef
Nasr, R, Lallemand-Breitenbach, V, Zhu, J, Guillemin, MC, de The, H. Therapy-induced PML/RARA proteolysis and acute promyelocytic leukemia cure. Clinical Cancer Research 2009;15:6321–6.CrossRef
Golub, TR, Slonim, DK, Tamayo, P, et al. Molecular classification of cancer: class discovery and class prediction by gene expression monitoring. Science 1999;286:531–7.CrossRef
Scholl, C, Bansal, D, Dohner, K, et al. The homeobox gene CDX2 is aberrantly expressed in most cases of acute myeloid leukemia and promotes leukemogenesis. Journal of Clinical Investigation 2007;117:1037–48.CrossRefGoogle ScholarPubMed
Nakamura, T, Largaespada, DA, Lee, MP, et al. Fusion of the nucleoporin gene NUP98 to HOXA9 by the chromosome translocation t(7;11)(p15;p15) in human myeloid leukaemia. Nature Genetics 1996;12:154–8.CrossRef
Borrow, J, Shearman, AM, Stanton, VP, Jr., et al. The t(7;11)(p15;p15) translocation in acute myeloid leukaemia fuses the genes for nucleoporin NUP98 and class I homeoprotein HOXA9. Nature Genetics 1996;12:159–67.CrossRef
Milne, TA, Briggs, SD, Brock, HW, et al. MLL targets SET domain methyltransferase activity to Hox gene promoters. Molecular Cell 2002;10:1107–17.CrossRef
Barry, ER, Corry, GN, Rasmussen, TP. Targeting DOT1L action and interactions in leukemia: the role of DOT1L in transformation and development. Expert Opinion on Therapeutic Targets 2010;14:405–18.CrossRef
Daigle, SR, Olhava, EJ, Therkelsen, CA, et al. Selective killing of mixed lineage leukemia cells by a potent small-molecule DOT1L inhibitor. Cancer Cell 2011;20:53–65.CrossRef
Kreidberg, JA, Sariola, H, Loring, JM, et al. WT-1 is required for early kidney development. Cell 1993;74:679–91.CrossRef
Ariyaratana, S, Loeb, DM. The role of the Wilms tumour gene (WT1) in normal and malignant haematopoiesis. Expert Reviews in Molecular Medicine 2007;9:1–17.CrossRef
Rezvani, K. Posttransplantation vaccination: concepts today and on the horizon. Hematology/The Education Program of the American Society of Hematology 2011:299–304.
Kohrt, HE, Muller, A, Baker, J, et al. Donor immunization with WT1 peptide augments antileukemic activity after MHC-matched bone marrow transplantation. Blood 2011;118:5319–29.CrossRef
Ochsenreither, S, Fusi, A, Busse, A, et al. “Wilms Tumor Protein 1” (WT1) peptide vaccination-induced complete remission in a patient with acute myeloid leukemia is accompanied by the emergence of a predominant T-cell clone both in blood and bone marrow. Journal of Immunotherapy 2011;34:85–91.CrossRefGoogle Scholar
Inoue, K, Sugiyama, H, Ogawa, H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood 1994;84:3071–9.
Rezvani, K, Yong, AS, Savani, BN, et al. Graft-versus-leukemia effects associated with detectable Wilms tumor-1 specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia. Blood 2007;110:1924–32.CrossRef
Gaidzik, VI, Schlenk, RF, Moschny, S, et al. Prognostic impact of WT1 mutations in cytogenetically normal acute myeloid leukemia: a study of the German-Austrian AML Study Group. Blood 2009;113:4505–11.CrossRef
Hou, HA, Huang, TC, Lin, LI, et al. WT1 mutation in 470 adult patients with acute myeloid leukemia: stability during disease evolution and implication of its incorporation into a survival scoring system. Blood 2010;115:5222–31.CrossRef
Yan, XJ, Xu, J, Gu, ZH, et al. Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia. Nature Genetics 2011;43:309–15.CrossRef
Paschka, P, Marcucci, G, Ruppert, AS, et al. Wilms’ tumor 1 gene mutations independently predict poor outcome in adults with cytogenetically normal acute myeloid leukemia: a cancer and leukemia group B study. Journal of Clinical Oncology 2008;26:4595–602.CrossRefGoogle ScholarPubMed
Ellisen, LW, Carlesso, N, Cheng, T, Scadden, DT, Haber, DA. The Wilms tumor suppressor WT1 directs stage-specific quiescence and differentiation of human hematopoietic progenitor cells. EMBO Journal 2001;20:1897–909.CrossRef
King-Underwood, L, Little, S, Baker, M, et al. Wt1 is not essential for hematopoiesis in the mouse. Leukemia Research 2005;29:803–12.CrossRef
Alberta, JA, Springett, GM, Rayburn, H, et al. Role of the WT1 tumor suppressor in murine hematopoiesis. Blood 2003;101:2570–4.CrossRef
Wagner, KJ, Patek, CE, Cunningham, A, et al. C-terminal truncation of WT1 delays but does not abolish hematopoiesis in embryoid bodies. Blood Cells, Molecules and Diseases 2002;28:428–35.CrossRef
Welch, JS, Yuan, W, Ley, TJ.PML-RARA can increase hematopoietic self-renewal without causing a myeloproliferative disease in mice. Journal of Clinical Investigation 2011;121:1636–45.CrossRefGoogle ScholarPubMed
Hosen, N, Shirakata, T, Nishida, S, et al. The Wilms’ tumor gene WT1-GFP knock-in mouse reveals the dynamic regulation of WT1 expression in normal and leukemic hematopoiesis. Leukemia 2007;21:1783–91.CrossRef
Nishida, S, Hosen, N, Shirakata, T, et al. AML1-ETO rapidly induces acute myeloblastic leukemia in cooperation with the Wilms tumor gene, WT1. Blood 2006;107:3303–12.CrossRef
Yamashita, Y, Yuan, J, Suetake, I, et al. Array-based genomic resequencing of human leukemia. Oncogene 2010;29:3723–31.CrossRef
Walter, MJ, Ding, L, Shen, D, et al. Recurrent DNMT3A mutations in patients with myelodysplastic syndromes. Leukemia 2011;25:1153–8.CrossRef
Stegelmann, F, Bullinger, L, Schlenk, RF, et al. DNMT3A mutations in myeloproliferative neoplasms. Leukemia 2011;25:1217–19.CrossRef
Thol, F, Damm, F, Ludeking, A, et al. Incidence and prognostic influence of DNMT3A mutations in acute myeloid leukemia. Journal of Clinical Oncology 2011;29:2889–96.CrossRefGoogle ScholarPubMed
Shen, Y, Zhu, YM, Fan, X, et al. Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia. Blood 2011;118:5593–603.CrossRef
Holz-Schietinger, C, Matje, DM, Harrison, MF, Reich NO. Oligomerization of DNMT3A controls the mechanism of de novo DNA methylation. Journal of Biological Chemistry 2011;286:41 479–88.CrossRefGoogle ScholarPubMed
Wu, H, Coskun, V, Tao, J, et al. Dnmt3a-dependent nonpromoter DNA methylation facilitates transcription of neurogenic genes. Science 2010;329:444–8.CrossRef
Blum, W, Garzon, R, Klisovic, RB, et al. Clinical response and miR-29b predictive significance in older AML patients treated with a 10-day schedule of decitabine. Proceedings of the National Academy of Sciences USA 2010;107:7473–8.CrossRef
Metzeler, KH, Walker, A, Geyer, S, et al. DNMT3A mutations and response to the hypomethylating agent decitabine in acute myeloid leukemia. Leukemia 2011;26:1106–7.CrossRef
Broske, AM, Vockentanz, L, Kharazi, S, et al. DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nature Genetics 2009;41:1207–15.CrossRef
Challen, GA, Sun, D, Jeong, M, et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nature Genetics 2012;44:23–31.CrossRef
Gao, Q, Steine, EJ, Barrasa, MI, et al. Deletion of the de novo DNA methyltransferase Dnmt3a promotes lung tumor progression. Proceedings of the National Academy of Sciences USA 2011;108:18 061–6.
Xu, Q, Simpson, SE, Scialla, TJ, Bagg, A, Carroll, M. Survival of acute myeloid leukemia cells requires PI3 kinase activation. Blood 2003;102:972–80.CrossRef
Campos, L, Rouault, JP, Sabido, O, et al. High expression of bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Blood 1993;81:3091–6.
Meek, DW. Tumour suppression by p53: a role for the DNA damage response? Nature Reviews Cancer 2009;9:714–23.
Drexler, HG. Review of alterations of the cyclin-dependent kinase inhibitor INK4 family genes p15, p16, p18 and p19 in human leukemia-lymphoma cells. Leukemia 1998;12:845–59.CrossRef
Chim, CS, Wong, AS, Kwong, YL. Epigenetic inactivation of INK4/CDK/RB cell cycle pathway in acute leukemias. Annals of Hematology 2003;82:738–42.CrossRef
Chim, CS, Wong, AS, Kwong, YL. Epigenetic inactivation of the CIP/KIP cell-cycle control pathway in acute leukemias. American Journal of Hematology 2005;80:282–7.CrossRefGoogle ScholarPubMed
Wang, L, Wang, J, Blaser, BW, et al. Pharmacologic inhibition of CDK4/6: mechanistic evidence for selective activity or acquired resistance in acute myeloid leukemia. Blood 2007;110:2075–83.CrossRef
Wattel, E, Preudhomme, C, Hecquet, B, et al. p53 mutations are associated with resistance to chemotherapy and short survival in hematologic malignancies. Blood 1994;84:3148–57.
Sallmyr, A, Fan, J, Datta, K, et al. Internal tandem duplication of FLT3 (FLT3/ITD) induces increased ROS production, DNA damage, and misrepair: implications for poor prognosis in AML. Blood 2008;111:3173–2.CrossRef
Lin, YH, Kakadia, PM, Chen, Y, et al. Global reduction of the epigenetic H3K79 methylation mark and increased chromosomal instability in CALM-AF10-positive leukemias. Blood 2009;114:651–8.CrossRef
Falini, B, Gionfriddo, I, Cecchetti, F, et al. Acute myeloid leukemia with mutated nucleophosmin (NPM1): any hope for a targeted therapy? Blood Reviews 2011;25:247–54.
Grisendi, S, Bernardi, R, Rossi, M, et al. Role of nucleophosmin in embryonic development and tumorigenesis. Nature 2005;437:147–53.CrossRef
Okuda, M, Horn, HF, Tarapore, P, et al. Nucleophosmin/B23 is a target of CDK2/cyclin E in centrosome duplication. Cell 2000;103:127–40.CrossRef
den Besten, W, Kuo, ML, Williams, RT, Sherr, CJ. Myeloid leukemia-associated nucleophosmin mutants perturb p53-dependent and independent activities of the Arf tumor suppressor protein. Cell Cycle 2005;4:1593–8.CrossRef
Colombo, E, Martinelli, P, Zamponi, R, et al. Delocalization and destabilization of the Arf tumor suppressor by the leukemia-associated NPM mutant. Cancer Research. 2006;66:3044–50.CrossRef
Sportoletti, P, Grisendi, S, Majid, SM, et al. Npm1 is a haploinsufficient suppressor of myeloid and lymphoid malignancies in the mouse. Blood 2008;111:3859–62.CrossRef
Cheng, K, Sportoletti, P, Ito, K, et al. The cytoplasmic NPM mutant induces myeloproliferation in a transgenic mouse model. Blood 2010;115:3341–5.CrossRef
Li, Z, Boone, D, Hann, SR. Nucleophosmin interacts directly with c-Myc and controls c-Myc-induced hyperproliferation and transformation. Proceedings of the National Academy of Sciences USA 2008;105:18 794–9.
Bonetti, P, Davoli, T, Sironi, C, et al. Nucleophosmin and its AML-associated mutant regulate c-Myc turnover through Fbw7 gamma. Journal of Cell Biology 2008;182:19–26.CrossRefGoogle ScholarPubMed
Leong, SM, Tan, BX, Ahmad, B, et al. Mutant nucleophosmin deregulates cell death and myeloid differentiation through excessive caspase-6 and -8 inhibition. Blood 2010;116:3286–96.CrossRef
Warburg, O. On respiratory impairment in cancer cells. Science 1956;124:269–70.
Parsons, DW, Jones, S, Zhang, X, et al. An integrated genomic analysis of human glioblastoma multiforme. Science 2008;321:1807–12.CrossRef
Yan, H, Parsons, DW, Jin, G, et al. IDH1 and IDH2 mutations in gliomas. New England Journal of Medicine. 2009;360:765–73.CrossRefGoogle ScholarPubMed
Gross, S, Cairns, RA, Minden, MD, et al. Cancer-associated metabolite 2-hydroxyglutarate accumulates in acute myelogenous leukemia with isocitrate dehydrogenase 1 and 2 mutations. Journal of Experimental Medicine 2010;207:339–44.CrossRefGoogle ScholarPubMed
Ward, PS, Patel, J, Wise, DR, et al. The common feature of leukemia-associated IDH1 and IDH2 mutations is a neomorphic enzyme activity converting alpha-ketoglutarate to 2-hydroxyglutarate. Cancer Cell 2010;17:225–34.CrossRef
Figueroa, ME, Abdel-Wahab, O, Lu, C, et al. Leukemic IDH1 and IDH2 mutations result in a hypermethylation phenotype, disrupt TET2 function, and impair hematopoietic differentiation. Cancer Cell 2010;18:553–67.CrossRef
He, YF, Li, BZ, Li, Z, et al. Tet-mediated formation of 5-carboxylcytosine and its excision by TDG in mammalian DNA. Science 2011;333:1303–7.CrossRef
Ito, S, Shen, L, Dai, Q, et al. Tet proteins can convert 5-methylcytosine to 5-formylcytosine and 5-carboxylcytosine. Science 2011;333:1300–3.CrossRef
Ko, M, Huang, Y, Jankowska, AM, et al. Impaired hydroxylation of 5-methylcytosine in myeloid cancers with mutant TET2. Nature 2010;468:839–43.CrossRef
Moran-Crusio, K, Reavie, L, Shih, A, et al. Tet2 loss leads to increased hematopoietic stem cell self-renewal and myeloid transformation. Cancer Cell 2011;20:11–24.CrossRef
Ko, M, Bandukwala, HS, An, J, et al. Ten-Eleven-Translocation 2 (TET2) negatively regulates homeostasis and differentiation of hematopoietic stem cells in mice. Proceedings of the National Academy of Sciences USA 2011;108:14 566–71.
Quivoron, C, Couronne, L, Della Valle, V, et al. TET2 inactivation results in pleiotropic hematopoietic abnormalities in mouse and is a recurrent event during human lymphomagenesis. Cancer Cell 2011;20:25–38.CrossRef
Abdel-Wahab, O, Mullally, A, Hedvat, C, et al. Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood 2009;114:144–7.CrossRef
Xu, W, Yang, H, Liu, Y, et al. Oncometabolite 2-hydroxyglutarate is a competitive inhibitor of alpha-ketoglutarate-dependent dioxygenases. Cancer Cell 2011;19:17–30.CrossRef
Lu, C, Ward, PS, Kapoor, GS, et al. IDH mutation impairs histone demethylation and results in a block to cell differentiation. Nature. 2012;183:474–8.
Zhao, S, Lin, Y, Xu, W, et al. Glioma-derived mutations in IDH1 dominantly inhibit IDH1 catalytic activity and induce HIF-1alpha. Science 2009;324:261–5.CrossRef
Takubo, K, Goda, N, Yamada, W, et al. Regulation of the HIF-1alpha level is essential for hematopoietic stem cells. Cell Stem Cell 2010;7:391–402.CrossRef
Kirito, K, Fox, N, Komatsu, N, Kaushansky, K. Thrombopoietin enhances expression of vascular endothelial growth factor (VEGF) in primitive hematopoietic cells through induction of HIF-1alpha. Blood 2005;105:4258–63.CrossRef
Pedersen, M, Lofstedt, T, Sun, J, et al. Stem cell factor induces HIF-1alpha at normoxia in hematopoietic cells. Biochemical and Biophysical Research Communications 2008;377:98–103.CrossRef
Simon, MC, Keith, B. The role of oxygen availability in embryonic development and stem cell function. Nature Reviews Molecular and Cellular Biology 2008;9:285–96.CrossRef
Weisberg, E, Manley, PW, Breitenstein, W, et al. Characterization of AMN107, a selective inhibitor of native and mutant Bcr-Abl. Cancer Cell 2005;7:129–41.CrossRef
Das, J, Chen, P, Norris, D, et al. 2-aminothiazole as a novel kinase inhibitor template. Structure-activity relationship studies toward the discovery of N-(2-chloro-6-methylphenyl)-2-[[6-[4-(2-hydroxyethyl)-1-piperazinyl)]-2-methyl-4-pyrimidinyl]amino)]-1,3-thiazole-5-carboxamide (dasatinib, BMS-354825) as a potent pan-Src kinase inhibitor. Journal of Medicinal Chemistry 2006;49:6819–32.CrossRefGoogle ScholarPubMed
Smith, BD, Levis, M, Beran, M, et al. Single-agent CEP-701, a novel FLT3 inhibitor, shows biologic and clinical activity in patients with relapsed or refractory acute myeloid leukemia. Blood 2004;103:3669–76.CrossRef
Knapper, S, Burnett, AK, Littlewood, T, et al. A phase 2 trial of the FLT3 inhibitor lestaurtinib (CEP701) as first-line treatment for older patients with acute myeloid leukemia not considered fit for intensive chemotherapy. Blood 2006;108:3262–70.CrossRef
O’Farrell, AM, Foran, JM, Fiedler, W, et al. An innovative Phase I clinical study demonstrates inhibition of FLT3 phosphorylation by SU11248 in acute myeloid leukemia patients. Clinical Cancer Research 2003;9:5465–76.
Fiedler, W, Serve, H, Dohner, H, et al. A phase 1 study of SU11248 in the treatment of patients with refractory or resistant acute myeloid leukemia (AML) or not amenable to conventional therapy for the disease. Blood 2005;105:986–93.CrossRef
Stone, RM, DeAngelo, DJ, Klimek, V, et al. Patients with acute myeloid leukemia and an activating mutation in FLT3 respond to a small-molecule FLT3 tyrosine kinase inhibitor, PKC412. Blood 2005;105:54–60.CrossRef
DeAngelo, DJ, Stone, RM, Heaney, ML, et al. Phase 1 clinical results with tandutinib (MLN518), a novel FLT3 antagonist, in patients with acute myelogenous leukemia or high-risk myelodysplastic syndrome: safety, pharmacokinetics, and pharmacodynamics. Blood 2006;108:3674–81.CrossRef
DeAngelo, DJ, Amrein, PC, Kovacsovics, TJ, et al. Phase 1/2 study of tandutinib (MLN518) plus standard induction chemotherapy in newly diagnosed acute myelogenous leukemia (AML). ASH Annual Meeting Abstracts 2006;108:158.
Weisberg, E, Boulton, C, Kelly, LM, et al. Inhibition of mutant FLT3 receptors in leukemia cells by the small molecule tyrosine kinase inhibitor PKC412. Cancer Cell 2002;1:433–43.CrossRef
Heidel, F, Solem, FK, Breitenbuecher, F, et al. Clinical resistance to the kinase inhibitor PKC412 in acute myeloid leukemia by mutation of Asn-676 in the FLT3 tyrosine kinase domain. Blood 2006;107:293–300.CrossRef
Weisberg, E, Choi, HG, Barrett, R, et al. Discovery and characterization of novel mutant FLT3 kinase inhibitors. Molecular Cancer Therapeutics 2010;9:2468–77.CrossRef
Karp, JE, Lancet, JE, Kaufmann, SH, et al. Clinical and biologic activity of the farnesyltransferase inhibitor R115777 in adults with refractory and relapsed acute leukemias: a phase 1 clinical-laboratory correlative trial. Blood 2001;97:3361–9.CrossRef
Lancet, JE, Gojo, I, Gotlib, J, et al. A phase 2 study of the farnesyltransferase inhibitor tipifarnib in poor-risk and elderly patients with previously untreated acute myelogenous leukemia. Blood 2007;109:1387–94.CrossRef
Tothova, E, Fricova, M, Stecova, N, Kafkova, A, Elbertova, A. High expression of Bcl-2 protein in acute myeloid leukemia cells is associated with poor response to chemotherapy. Neoplasma 2002;49:141–4.
Marcucci, G, Stock, W, Dai, G, et al. Phase I study of oblimersen sodium, an antisense to Bcl-2, in untreated older patients with acute myeloid leukemia: pharmacokinetics, pharmacodynamics, and clinical activity. Journal of Clinical Oncology 2005;23:3404–11.CrossRefGoogle ScholarPubMed
Oltersdorf, T, Elmore, SW, Shoemaker, AR, et al. An inhibitor of Bcl-2 family proteins induces regression of solid tumours. Nature 2005;435:677–81.CrossRef
LaCasse, EC, Cherton-Horvat, GG, Hewitt, KE, et al. Preclinical characterization of AEG35156/GEM 640, a second-generation antisense oligonucleotide targeting X-linked inhibitor of apoptosis. Clinical Cancer Research 2006;12:5231–41.CrossRef
Wuchter, C, Krappmann, D, Cai, Z, et al. In vitro susceptibility to TRAIL-induced apoptosis of acute leukemia cells in the context of TRAIL receptor gene expression and constitutive NF-kappa B activity. Leukemia 2001;15:921–8.CrossRef
Suh, WS, Kim, YS, Schimmer, AD, et al. Synthetic triterpenoids activate a pathway for apoptosis in AML cells involving downregulation of FLIP and sensitization to TRAIL. Leukemia 2003;17:2122–9.CrossRef
Mathews, V, George, B, Lakshmi, KM, et al. Single-agent arsenic trioxide in the treatment of newly diagnosed acute promyelocytic leukemia: durable remissions with minimal toxicity. Blood 2006;107:2627–32.CrossRef
Ghavamzadeh, A, Alimoghaddam, K, Ghaffari, SH, et al. Treatment of acute promyelocytic leukemia with arsenic trioxide without ATRA and/or chemotherapy. Annals of Oncology 2006;17:131–4.CrossRef
Jing, Y, Wang, L, Xia, L, et al. Combined effect of all-trans retinoic acid and arsenic trioxide in acute promyelocytic leukemia cells in vitro and in vivo. Blood 2001;97:264–9.CrossRef
Shackelford, D, Kenific, C, Blusztajn, A, Waxman, S, Ren, R. Targeted degradation of the AML1/MDS1/EVI1 oncoprotein by arsenic trioxide. Cancer Research 2006;66:11 360–9.
Linenberger, ML. CD33-directed therapy with gemtuzumab ozogamicin in acute myeloid leukemia: progress in understanding cytotoxicity and potential mechanisms of drug resistance. Leukemia 2005;19:176–82.CrossRef
Feuring-Buske, M, Frankel, AE, Alexander, RL, Gerhard, B, Hogge, DE. A diphtheria toxin-interleukin 3 fusion protein is cytotoxic to primitive acute myeloid leukemia progenitors but spares normal progenitors. Cancer Research 2002;62:1730–6.
Gallay, N, Anani, L, Lopez, A, et al. The role of platelet/endothelial cell adhesion molecule 1 (CD31) and CD38 antigens in marrow microenvironmental retention of acute myelogenous leukemia cells. Cancer Research 2007;67:8624–32.CrossRef
Nervi, B, Ramirez, P, Rettig, MP, et al. Chemosensitization of acute myeloid leukemia (AML) following mobilization by the CXCR4 antagonist AMD3100. Blood 2009;113:6206–14.CrossRef
Deshpande, AJ, Cusan, M, Rawat, VP, et al. Acute myeloid leukemia is propagated by a leukemic stem cell with lymphoid characteristics in a mouse model of CALM/AF10-positive leukemia. Cancer Cell 2006;10:363–74.CrossRef
van Rhenen, A, Moshaver, B, Kelder, A, et al. Aberrant marker expression patterns on the CD34+CD38- stem cell compartment in acute myeloid leukemia allows to distinguish the malignant from the normal stem cell compartment both at diagnosis and in remission. Leukemia 2007;21:1700–7.CrossRef
Bakker, AB, van den Oudenrijn, S, Bakker, AQ, et al. C-type lectin-like molecule-1: a novel myeloid cell-surface marker associated with acute myeloid leukemia. Cancer Research 2004;64:8443–50.CrossRef
Tavor, S, Petit, I, Porozov, S, et al. CXCR4 regulates migration and development of human acute myelogenous leukemia stem cells in transplanted NOD/SCID mice. Cancer Research 2004;64:2817–24.CrossRef
Rombouts, EJ, Pavic, B, Lowenberg, B, Ploemacher, RE. Relation between CXCR-4 expression, Flt3 mutations, and unfavorable prognosis of adult acute myeloid leukemia. Blood 2004;104:550–7.CrossRef
Zeng, Z, Samudio, IJ, Munsell, M, et al. Inhibition of CXCR4 with the novel RCP168 peptide overcomes stroma-mediated chemoresistance in chronic and acute leukemias. Molecular Cancer Therapeutics 2006;5:3113–21.CrossRef
Krause, DS, Lazarides, K, von Andrian, UH, Van Etten RA. Requirement for CD44 in homing and engraftment of BCR-ABL-expressing leukemic stem cells. Nature Medicine 2006;12:1175–80.CrossRef
Nishioka, C, Ikezoe, T, Yang, J, et al. Blockade of MEK/ERK signaling enhances sunitinib-induced growth inhibition and apoptosis of leukemia cells possessing activating mutations of the FLT3 gene. Leukemia Research 2008;32:865–72.CrossRef

Save book to Kindle

To save this book to your Kindle, first ensure coreplatform@cambridge.org is added to your Approved Personal Document E-mail List under your Personal Document Settings on the Manage Your Content and Devices page of your Amazon account. Then enter the ‘name’ part of your Kindle email address below. Find out more about saving to your Kindle.

Note you can select to save to either the @free.kindle.com or @kindle.com variations. ‘@free.kindle.com’ emails are free but can only be saved to your device when it is connected to wi-fi. ‘@kindle.com’ emails can be delivered even when you are not connected to wi-fi, but note that service fees apply.

Find out more about the Kindle Personal Document Service.

  • The molecular basis of acute myeloid leukemia
    • By Kim L. Rice, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Monica Buzzai, Novartis, Origgio, VA, Italy, Jessica Altman, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Jonathan D. Licht, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.070
Available formats
×

Save book to Dropbox

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Dropbox.

  • The molecular basis of acute myeloid leukemia
    • By Kim L. Rice, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Monica Buzzai, Novartis, Origgio, VA, Italy, Jessica Altman, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Jonathan D. Licht, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.070
Available formats
×

Save book to Google Drive

To save content items to your account, please confirm that you agree to abide by our usage policies. If this is the first time you use this feature, you will be asked to authorise Cambridge Core to connect with your account. Find out more about saving content to Google Drive.

  • The molecular basis of acute myeloid leukemia
    • By Kim L. Rice, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Monica Buzzai, Novartis, Origgio, VA, Italy, Jessica Altman, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA, Jonathan D. Licht, Division of Hematology/Oncology, Robert H. Lurie Comprehensive Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
  • Edited by Edward P. Gelmann, Columbia University, New York, Charles L. Sawyers, Memorial Sloan-Kettering Cancer Center, New York, Frank J. Rauscher, III
  • Book: Molecular Oncology
  • Online publication: 05 February 2015
  • Chapter DOI: https://doi.org/10.1017/CBO9781139046947.070
Available formats
×