Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-qxdb6 Total loading time: 0 Render date: 2024-04-27T11:19:25.038Z Has data issue: false hasContentIssue false

8 - Acute myeloid leukemia

from Part 2 - Hematological malignancies

Published online by Cambridge University Press:  06 December 2010

Wendy N. Erber
Affiliation:
University of Western Australia, Perth
Get access

Summary

Introduction

Acute myeloid leukemia (AML) encompasses a highly heterogeneous group of clonal disorders arising in hematopoietic progenitors that are characterized by a block in differentiation and outgrowth of myeloid blasts giving rise to bone marrow failure. Although AML is not particularly common, affecting approximately 3 individuals per 100 000 population per year in Western countries (i.e. ~2000 new cases per year in the UK), it is challenging and expensive to treat, representing a significant burden on healthcare systems. Given that AML is predominantly a disease of the elderly, with markedly higher incidence in individuals over 60 years of age, this disease is set to become an increasing problem as the population ages.

Molecular basis of AML

The last three decades have witnessed major advances in deciphering the cytogenetic and molecular lesions underlying the pathogenesis of AML. These have not only afforded significant insights into disease biology, but also proved helpful in providing prognostic information and underpinned the development of molecularly targeted and risk-stratified treatment approaches. A further benefit of improved understanding of the molecular basis of AML coupled with the development of sensitive quantitative polymerase chain techniques has been the possibility to assess treatment response at the submicroscopic level (i.e. detection of minimal residual disease, MRD), thereby affording the opportunity to tailor therapy more precisely to the needs of the individual patient.

Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2010

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

Grimwade, D. Impact of cytogenetics on clinical outcome in AML. In Karp, JE (ed.), Acute Myelogenous Leukemia. Totowa, NJ: Humana Press; 2007: 177–92.CrossRefGoogle Scholar
,Mitelman Database of Chromosome Aberrations in Cancer (2009). Mitelman, F, Johansson, B, Mertens, F (eds.), http://cgap.nci.nih.gov/Chromosomes/Mitelman
Rosenbauer, F, Tenen, DG. Transcription factors in myeloid development: balancing differentiation with transformation. Nat Rev Immunol 2007;7:105–17.CrossRefGoogle ScholarPubMed
Grimwade, D, Mistry, AR, Solomon, E, Guidez, F. Acute promyelocytic leukemia: A paradigm for differentiation therapy. Cancer Treat Res, 2009;145: 219–35CrossRefGoogle Scholar
Lugthart, S, Drunen, E, Norden, Yet al. High EVI1 levels predict adverse outcome in acute myeloid leukemia: prevalence of EVI1 overexpression and chromosome 3q26 abnormalities underestimated. Blood 2008;111:4329–37.CrossRefGoogle ScholarPubMed
Guidez, F, Zelent, A. Role of nuclear receptor corepressors in leukemogenesis. Curr Top Microbiol Immunol 2001;254:165–85.Google ScholarPubMed
Plass, C, Oakes, C, Blum, W, Marcucci, G.Epigenetics in acute myeloid leukemia. Semin Oncol 2008;35:378–87.CrossRefGoogle ScholarPubMed
Harper, DP, Aplan, PD. Chromosomal rearrangements leading to MLL gene fusions: clinical and biological aspects. Cancer Res 2008;68:10024–7.CrossRefGoogle ScholarPubMed
Moore, MA, Chung, KY, Plasilova, Met al. NUP98dysregulation in myeloid leukemogenesis. Ann N Y Acad Sci 2007;1106:114–42.CrossRefGoogle ScholarPubMed
Caligiuri, MA, Strout, MP, Oberkircher, ARet al. The partial tandem duplication of ALL1 in acute myeloid leukemia with normal cytogenetics or trisomy 11 is restricted to one chromosome. Proc Natl Acad Sci USA 1997;94:3899–902.CrossRefGoogle ScholarPubMed
Liu, TX, Becker, MW, Jelinek, Jet al. Chromosome 5q deletion and epigenetic suppression of the gene encoding alpha-catenin (CTNNA1) in myeloid cell transformation. Nat Med 2007;13:78–83.CrossRefGoogle Scholar
Ebert, BL, Pretz, J, Bosco, Jet al. Identification of RPS14 as a 5q- syndrome gene by RNA interference screen. Nature 2008;451:335–9.CrossRefGoogle ScholarPubMed
Ebert, BL. Deletion 5q in myelodysplastic syndrome: a paradigm for the study of hemizygous deletions in cancer. Leukemia 2009;23:1252–6.CrossRefGoogle Scholar
Mrózek, K, Marcucci, G, Paschka, P, Whitman, SP, Bloomfield, CD. Clinical relevance of mutations and gene-expression changes in adult acute myeloid leukemia with normal cytogenetics: are we ready for a prognostically prioritized molecular classification?Blood 2007;109:431–48.CrossRefGoogle ScholarPubMed
Scholl, C, Gilliland, DG, Fröhling, S. Deregulation of signaling pathways in acute myeloid leukemia. Semin Oncol 2008;35:336–45.CrossRefGoogle ScholarPubMed
Mead, AJ, Linch, DC, Hills, RKet al. FLT3 tyrosine kinase domain mutations are biologically distinct from and have a significantly more favorable prognosis than FLT3 internal tandem duplications in patients with acute myeloid leukemia. Blood 2007;110:1262–70.CrossRefGoogle ScholarPubMed
Bacher, U, Haferlach, C, Kern, Wet al. Prognostic relevance of FLT3-TKD mutations in AML: the combination matters – an analysis of 3082 patients. Blood 2008;111:2527–37.CrossRefGoogle ScholarPubMed
Whitman, SP, Ruppert, AS, Radmacher, MDet al. FLT3 D835/I836 mutations are associated with poor disease-free survival and a distinct gene-expression signature among younger adults with de novo cytogenetically normal acute myeloid leukemia lacking FLT3 internal tandem duplications. Blood 2008;111:1552–9.CrossRefGoogle Scholar
Whitman, SP, Archer, KJ, Feng, Let al. Absence of the wild-type allele predicts poor prognosis in adult de novo acute myeloid leukemia with normal cytogenetics and the internal tandem duplication of FLT3: a Cancer and Leukemia Group B study. Cancer Res 2001;61:7233–9.Google ScholarPubMed
Thiede, C, Steudel, C, Mohr, Bet al. Analysis of FLT3-activating mutations in 979 patients with acute myelogenous leukemia: association with FAB subtypes and identification of subgroups with poor prognosis. Blood 2002;99:4326–35.CrossRefGoogle ScholarPubMed
Gale, RE, Green, C, Allen, Cet 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.CrossRefGoogle Scholar
Fitzgibbon, J, Smith, LL, Raghavan, Met al. Association between acquired uniparental disomy and homozygous gene mutation in acute myeloid leukemias. Cancer Res 2005;65:9152–4.CrossRefGoogle ScholarPubMed
Falini, B, Mecucci, C, Tiacci, Eet al. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. New Engl J Med 2005;352:254–66.CrossRefGoogle ScholarPubMed
Falini, B, Nicoletti, I, Martelli, MF, Mecucci, C. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood 2007;109:874–85.CrossRefGoogle ScholarPubMed
Haferlach, C, Mecucci, C, Schnittger, Set al. AML with mutated NPM1 carrying a normal or aberrant karyotype show overlapping biological, pathological, immunophenotypic, and prognostic features. Blood 2009;114:3024–32.CrossRefGoogle ScholarPubMed
Pabst, T, Eyholzer, M, Haefliger, Set al. Somatic CEBPA mutations are a frequent second event in families with germline CEBPA mutations and familial acute myeloid leukemia. J Clin Oncol 2008;26:5088–93.CrossRefGoogle ScholarPubMed
Figueroa, ME, Wouters, BJ, Skrabanek, Let al. Genome-wide epigenetic analysis delineates a biologically distinct immature acute leukemia with myeloid/T-lymphoid features. Blood 2009;113:2795–804.CrossRefGoogle ScholarPubMed
Schlenk, RF, Döhner, K, Krauter, Jet al. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. New Engl J Med 2008;358:1909–18.CrossRefGoogle ScholarPubMed
Marcucci, G, Maharry, K, Radmacher, MDet al. Prognostic significance of, and gene and microRNA expression signatures associated with, CEBPA mutations in cytogenetically normal acute myeloid leukemia with high-risk molecular features: a Cancer and Leukemia Group B Study. J Clin Oncol 2008;26:5078–87.CrossRefGoogle ScholarPubMed
Wouters, BJ, Löwenberg, B, Erpelinck-Verschueren, CAet 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.CrossRefGoogle Scholar
Green, CL, Koo, KK, Hills, RKet 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. J Clin Oncol 2010;28:2739–47.CrossRefGoogle Scholar
Grimwade, D, Hills, RK. Independent prognostic factors for AML outcome. Hematology Am Soc Hematol Educ Program 2009; 385–95.Google ScholarPubMed
Abdel-Wahab, O, Mullally, A, Hedvat, Cet al. Genetic characterization of TET1, TET2, and TET3 alterations in myeloid malignancies. Blood 2009;114:144–7.CrossRefGoogle ScholarPubMed
Delhommeau, F, Dupont, S, Della Valle, Vet al. Mutation in TET2 in myeloid cancers. New Engl J Med 2009;360:2289–301.CrossRefGoogle ScholarPubMed
Mrózek, K, Marcucci, G, Paschka, P, Bloomfield, CD. Advances in molecular genetics and treatment of core-binding factor acute myeloid leukemia. Curr Opin Oncol 2008;20:711–18.CrossRefGoogle ScholarPubMed
Swerdlow, SH, Campo, E, Harris, NLet al. (eds.). World Health Organization Classification of Tumours of Haematopoietic and Lymphoid Tissues, 4th edn. Lyon: IARC Press; 2008.Google Scholar
Owen, C, Barnett, M, Fitzgibbon, J. Familial myelodysplasia and acute myeloid leukaemia – a review. Br J Haematol 2008;140:123–32.CrossRefGoogle ScholarPubMed
Dokal, I, Vulliamy, T. Inherited aplastic anaemias/bone marrow failure syndromes. Blood Rev 2008;22:141–53.CrossRefGoogle ScholarPubMed
Winter, JP, Joenje, H. The genetic and molecular basis of Fanconi anemia. Mutat Res 2009;668:11–19.CrossRefGoogle ScholarPubMed
Estey, E, Döhner, H. Acute myeloid leukaemia. Lancet 2006;368:1894–907.CrossRefGoogle ScholarPubMed
Godley, , Larson, RA. Therapy-related myeloid leukemia. Semin Oncol 2008;35:418–29.CrossRefGoogle ScholarPubMed
Bennett, JM, Catovsky, D, Daniel, MTet al. Proposed revised criteria for the classification of acute myeloid leukemia. A report of the French-American-British Cooperative Group. Ann Intern Med 1985;103:620–5.CrossRefGoogle ScholarPubMed
Jaffe, ES, Harris, NL, Stein, H, Vardiman, JW (eds.). World Health Organization Classification of Tumours: Pathology and Genetics of Tumours of Haematopoietic and Lymphoid Tissues. Lyon: IARC Press; 2001.
Milligan, DW, Grimwade, D, Cullis, JOet al. Guidelines on the management of acute myeloid leukaemia in adults. Br J Haematol 2006;135:450–74.Google ScholarPubMed
Sanz, MA, Grimwade, D, Tallman, MSet al. Management of acute promyelocytic leukemia: recommendations from an expert panel on behalf of the European LeukemiaNet. Blood 2009;113:1875–91.CrossRefGoogle Scholar
Döhner, H, Estey, EH, Amadori, Set al. Diagnosis and management of acute myeloid leukemia in adults: recommendations from an international expert panel, on behalf of the European LeukemiaNet. Blood 2010;115:453–74.CrossRefGoogle Scholar
Freeman, SD, Jovanovic, JV, Grimwade, D. Development of minimal residual disease directed therapy in acute myeloid leukemia. Semin Oncol 2008;35:388–400.CrossRefGoogle ScholarPubMed
Pinto, FO, Leblanc, T, Chamousset, Det al. Diagnosis of Fanconi anemia in patients with bone marrow failure. Haematologica 2009;94:487–95.CrossRefGoogle ScholarPubMed
Dongen, JJM, Macintyre, EA, Gabert, JAet al. Standardized RT-PCR analysis of fusion gene transcripts from chromosome aberrations in acute leukemia for detection of minimal residual disease. Leukemia 1999;13:1901–28.CrossRefGoogle ScholarPubMed
Metzgeroth, G, Walz, C, Score, Jet al. Recurrent finding of the FIP1L1-PDGFRA fusion gene in eosinophilia-associated acute myeloid leukemia and lymphoblastic T-cell lymphoma. Leukemia 2007;21:1183–8.CrossRefGoogle ScholarPubMed
Gulley, ML, Shea, TC, Fedoriw, Y. Genetic tests to evaluate prognosis and predict therapeutic response in acute myeloid leukemia. J Mol Diagn 2010;12:3–16.CrossRefGoogle ScholarPubMed
Schnittger, S, Kern, W, Tschulik, Cet al. Minimal residual disease levels assessed by NPM1 mutation specific RQ-PCR provide important prognostic information in AML. Blood 2009;114:2220–31.CrossRefGoogle ScholarPubMed
Schlenk, RF, Döhner, K, Kneba, Met al. Gene mutations and response to treatment with all-trans retinoic acid in elderly patients with acute myeloid leukemia. Results from the AMLSG Trial AML HD98B. Haematologica 2009;94:54–60.CrossRefGoogle ScholarPubMed
Rhenen, A, Feller, N, Kelder, Aet al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin Cancer Res 2005;11:6520–7.CrossRefGoogle ScholarPubMed
Lane, SW, Scadden, DT, Gilliland, DG. The leukemic stem cell niche – current concepts and therapeutic opportunities. Blood 2009;114:1150–7.CrossRefGoogle ScholarPubMed
Wouters, BJ, Löwenberg, B, Delwel, R. A decade of genome-wide gene expression profiling in acute myeloid leukemia: flashback and prospects. Blood 2009;113:291–8.CrossRefGoogle ScholarPubMed
Cheson, BD, Bennett, JM, Kopecky, KJet al. Revised recommendations of the International Working Group for Diagnosis, Standardization of Response Criteria, Treatment Outcomes, and Reporting Standards for Therapeutic Trials in Acute Myeloid Leukemia. J Clin Oncol 2003;21:4642–9.CrossRefGoogle Scholar
Gabert, JA, Beillard, E, Velden, VHJet al. Standardization and quality control studies of ‘real-time’ quantitative reverse transcriptase polymerase chain reaction of fusion gene transcripts for residual disease detection in leukemia – a Europe Against Cancer program. Leukemia 2003;17:2318–57.CrossRefGoogle ScholarPubMed
Beillard, E, Pallisgaard, N, Velden, VHJet al. Evaluation of candidate control genes for diagnosis and residual disease detection in leukemic patients using ‘real-time’ quantitative reverse-transcriptase polymerase chain reaction (RQ-PCR) – a Europe Against Cancer program. Leukemia 2003;17:2474–86.CrossRefGoogle ScholarPubMed
Cilloni, D, Renneville, A, Hermitte, Fet al. Real-time quantitative PCR detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: A European LeukemiaNet study. J Clin Oncol 2009;27:5195–201.CrossRefGoogle ScholarPubMed
Maurillo, L, Buccisano, F, Del Principe, MIet al. Toward optimization of postremission therapy for residual disease-positive patients with acute myeloid leukemia. J Clin Oncol 2008;26:4944–51.CrossRefGoogle ScholarPubMed
Grimwade, D, Jovanovic, JV, Hills, RKet al. Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy. J Clin Oncol 2009;27:3650–8.CrossRefGoogle ScholarPubMed
Craddock, C, Tauro, S, Moss, P, Grimwade, D. Biology and management of relapsed acute myeloid leukaemia. Br J Haematol 2005;129:18–34.CrossRefGoogle ScholarPubMed
Bacher, U, Zander, AR, Haferlach, Tet al. Minimal residual disease diagnostics in myeloid malignancies in the post transplant period. Bone Marrow Transplant 2008;42:145–57.CrossRefGoogle ScholarPubMed
Grimwade, D, Hills, RK, Moorman, AVet 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.CrossRefGoogle ScholarPubMed
Rubnitz, JE, Inaba, H, Dhal, Get al. Minimal residual dieases-directed theropy for childhood acute myeloid leukemia: results of the AML02 multicentre trial. Lancet Oncol 2010;11:543–52.CrossRefGoogle Scholar
Corbacioglu, A, Scholl, C, Schlenk, RFet al. Prognostic impact of minimal residual dieases in CBFB-MYH11-positive acute myeloid leukemia. J Clin Oncol 2010 Jul 12 [Epub ahead of print].

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.

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.

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.

Available formats
×