Skip to main content Accessibility help
×
Hostname: page-component-76fb5796d-45l2p Total loading time: 0 Render date: 2024-04-25T11:05:15.759Z Has data issue: false hasContentIssue false

11 - Minimal Residual Disease in Acute Myeloid Leukaemia

Published online by Cambridge University Press:  01 February 2018

Anna Porwit
Affiliation:
Lunds Universitet, Sweden
Marie Christine Béné
Affiliation:
Université de Nantes, France
Get access

Summary

Image of the first page of this content. For PDF version, please use the ‘Save PDF’ preceeding this image.'
Type
Chapter
Information
Publisher: Cambridge University Press
Print publication year: 2018

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

Van Stijn, A., Kok, A., van der Pol, M.A., et al. A flow cytometric method to detect apoptosis-related protein expression in minimal residual disease in acute myeloid leukemia. Leukemia, 17 (2003), 780–6.CrossRefGoogle ScholarPubMed
Te Boekhorst, P.A., de Leeuw, K., Schoester, M., et al. Predominance of functional multidrug resistance (MDR-1) phenotype in CD34+ acute myeloid leukemia cells. Blood, 8 (1993), 3157–62.Google Scholar
Suárez, L., Vidriales, M-B., García-Laraña, J., et al. CD34+ cells from acute myeloid leukemia, myelodysplastic syndromes, and normal BM display different apoptosis and drug resistance-associated phenotypes. Clin Cancer Res, 10 (2004), 7599–606.CrossRefGoogle ScholarPubMed
Van der Pol, M.A., Feller, N., Ossenkoppele, G.J., et al. Minimal residual disease in acute myeloid leukemia is predicted by P-glycoprotein activity but not by multidrug resistance protein activity at diagnosis. Leukemia, 17 (2003), 1674–7.CrossRefGoogle Scholar
Terstappen, L.W., Safford, M., Unterhalt, M., et al. Flow cytometric characterization of acute myeloid leukemia, IV. Comparison to the differentiation pathway of normal hematopoietic progenitor cells. Leukemia, 6 (1992), 9931000.Google Scholar
Loken, M.R., Alonzo, T.A., Pardo, L., et al. Residual disease detected by multidimensional flow cytometry signifies high relapse risk in patients with de novo acute myeloid leukemia, a report from Children's Oncology Group. Blood, 120 (2012), 1581–8.CrossRefGoogle ScholarPubMed
Martens, A.C., Schultz, F.W. and Hagenbeek, A.. Nonhomogeneous distribution of leukemia in the BM during minimal residual disease. Blood, 70 (1987), 1073–8.CrossRefGoogle Scholar
Ossenkoppele, G. and Schuurhuis, G.J.. MRD in AML, time for redefinition of CR? Blood, 121 (2013), 2166–8.CrossRefGoogle ScholarPubMed
San Miguel, J.F., Martínez, A., Macedo, A., et al. Immunophenotyping investigation of minimal residual disease is a useful approach for predicting relapse in acute myeloid leukemia patients. Blood, 90 (1997), 2465–70.CrossRefGoogle ScholarPubMed
Feller, N., van der Pol, M.A., van Stijn, A., et al. MRD parameters using immunophenotypic detection methods are highly reliable in predicting survival in acute myeloid leukaemia. Leukemia, 18 (2004), 1380–90.CrossRefGoogle ScholarPubMed
Kern, W., Voskova, D., Schoch, C.. Determination of relapse risk based on assessment of minimal residual disease during complete remission by multiparameter flow cytometry in unselected patients with acute myeloid leukemia. Blood, 104 (2004), 3078–85.Google ScholarPubMed
San Miguel, J.F., Vidriales, M-B., López-Berges, C., et al. Early immunophenotypical evaluation of minimal residual disease in acute myeloid leukemia identifies different patient risk groups and may contribute to postinduction treatment stratification. Blood, 98 (2001), 1746–51.CrossRefGoogle ScholarPubMed
Venditti, A.F., Buccisano, F., Del Poeta, G., et al. Level of minimal residual disease after consolidation therapy predicts outcome in acute myeloid leukemia. Blood, 96 (2000), 3948–52.CrossRefGoogle ScholarPubMed
Terwijn, M., van Putten, W.J.L., Kelder, A., et al. High prognostic impact of flow cytometric minimal residual disease detection in acute myeloid leukemia, data from the HOVON/SAKK AML 42A study. J Clin Oncol, 31 (2013), 3889–97.CrossRefGoogle ScholarPubMed
Freeman, S.D., Virgo, P., Couzens, S., et al. Prognostic relevance of treatment response measured by flow cytometric residual disease detection in older patients with acute myeloid leukemia. J Clin Oncol, 31 (2013), 4123–31.CrossRefGoogle ScholarPubMed
Jourdan, E., Boissel, N., Chevret, S., et al. Prospective evaluation of gene mutations and minimal residual disease in patients with core binding factor acute myeloid leukemia. Blood, 121 (2013), 2213–24.CrossRefGoogle ScholarPubMed
Inaba, H., Coustan-Smith, E., Cao, X., et al. Comparative analysis of different approaches to measure treatment response in acute myeloid leukemia. J Clin Oncol, 30 (2012), 3625–32.CrossRefGoogle ScholarPubMed
Krönke, J., Schlenk, R.F., Jensen, K-O., et al. Monitoring of minimal residual disease in NPM1-mutated acute myeloid leukemia, a study from the German-Austrian acute myeloid leukemia study group. J Clin Oncol, 29 (2011), 2709–16.CrossRefGoogle ScholarPubMed
Schnittger, S., Kern, W., Tschulik, C., et al. Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Blood, 114 (2009), 2220–31.CrossRefGoogle ScholarPubMed
Cilloni, D., Renneville, A., Hermitte, F., et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia, a European LeukemiaNet study. J Clin Oncol, 27 (2009), 5195–201.CrossRefGoogle ScholarPubMed
Zeijlemaker, W., Schuurhuis, G.J.. Minimal Residual Disease and Leukemic Stem Cells in Acute Myeloid Leukemia. Edited by: Guenova, Margarita and Balatzenko, Gueorgui. Leukemia In Tech Open (Rijeka, Croatia, 2013), 132, ISBN 978-953-51-1127-6.Google Scholar
Schnittger, S. 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. Blood, 100 (2002), 5966.CrossRefGoogle ScholarPubMed
Kottaridis, P.D., Gale, R.E., Langabeer, S.E., 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, 100 (2002), 2393–8.CrossRefGoogle ScholarPubMed
Cloos, J., Goemans, B.F., Hess, C.J., et al. Stability and prognostic influence of FLT3 mutations in paired initial and relapsed AML samples. Leukemia, 20 (2006), 1217–20.CrossRefGoogle ScholarPubMed
Al-Mawali, A., Gillis, D. and Lewis, I.. The use of receiver operating characteristic analysis for detection of minimal residual disease using five-color multiparameter flow cytometry in acute myeloid leukemia identifies patients with high risk of relapse. Cytometry B Clin Cytom, 76 (2009), 91101.CrossRefGoogle ScholarPubMed
Cui, W., Zhang, D., Cunningham, M.T., et al. Leukemia-associated aberrant immunophenotype in patients with acute myeloid leukemia, changes at refractory disease or first relapse and clinicopathological findings. Int J Lab Hematol, 36 (2014), 636–49.CrossRefGoogle ScholarPubMed
Voskova, D., Schoch, C., Schnittger, S., et al. Stability of leukemia-associated aberrant immunophenotypes in patients with acute myeloid leukemia between diagnosis and relapse, comparison with cytomorphologic, cytogenetic, and molecular genetic findings. Cytometry B Clin Cytom, 62 (2004), 2538.CrossRefGoogle ScholarPubMed
Van der Velden, V.H.J., van der Sluijs-Geling, A., Gibson, B.E.S., et al. Clinical significance of flowcytometric minimal residual disease detection in pediatric acute myeloid leukemia patients treated according to the DCOG ANLL97/MRC AML12 protocol. Leukemia, 24 (2010), 1599–606.CrossRefGoogle Scholar
Buccisano, F., Maurillo, L., Gattei, V., et al. The kinetics of reduction of minimal residual disease impacts on duration of response and survival of patients with acute myeloid leukemia. Leukemia 20 (2006), 1783–9.CrossRefGoogle ScholarPubMed
Maurillo, L., Buccisano, F., Principe, M.I. Del, et al. Toward optimization of postremission therapy for residual disease-positive patients with acute myeloid leukemia. J Clin Oncol, 26 (2008), 4944–51.CrossRefGoogle ScholarPubMed
Brooimans, R.A., Kraan, J., van Putten, W., et al. Flow cytometric differential of leukocyte populations in normal BM, influence of PB contamination. Cytometry B Clin Cytom, 76 (2009), 1826.CrossRefGoogle Scholar
Feller, N., van der Velden, V.H.J., Brooimans, R.A., et al. Defining consensus leukemia-associated immunophenotypes for detection of minimal residual disease in acute myeloid leukemia in a multicenter setting. Blood Cancer J, 3 (2013), e129.CrossRefGoogle Scholar
Terwijn, M., Kelder, A., Snel, A.N., et al. Minimal residual disease detection defined as the malignant fraction of the total primitive stem cell compartment offers additional prognostic information in acute myeloid leukaemia. Int J Lab Hematol, 34 (2012), 432–41.CrossRefGoogle ScholarPubMed
Voskova, D., Schnittger, S., Schoch, C., et al. Use of five-color staining improves the sensitivity of multiparameter flow cytomeric assessment of minimal residual disease in patients with acute myeloid leukemia. Leuk Lymphoma, 48 (2007), 80–8.CrossRefGoogle ScholarPubMed
Feller, N., Schuurhuis, G. J., van der Pol, M.A., et al. High percentage of CD34-positive cells in autologous AML PB stem cell products reflects inadequate in vivo purging and low chemotherapeutic toxicity in a subgroup of patients with poor clinical outcome. Leukemia, 17 (2003), 6875.CrossRefGoogle Scholar
Van der Pol, M.A., Feller, N., Roseboom, M., et al. Assessment of the normal or leukemic nature of CD34+ cells in acute myeloid leukemia with low percentages of CD34 cells. Haematologica, 88 (2003), 983–93.Google ScholarPubMed
Van Stijn, A., Kok, A., van Stalborch, M.A., et al. Minimal residual disease cells in AML patients have an apoptosis-sensitive protein profile. Leukemia, 18 (2004), 875–7.CrossRefGoogle ScholarPubMed
Kalina, T., Flores-Montero, J., van der Velden, V.H.J., et al. EuroFlow standardization of flow cytometer instrument settings and immunophenotyping protocols. Leukemia, 26 (2012), 19862010.CrossRefGoogle ScholarPubMed
Van Dongen, J.J.M., Lhermitte, L., Böttcher, S., et al. EuroFlow antibody panels for standardized n-dimensional flow cytometric immunophenotyping of normal, reactive and malignant leukocytes. Leukemia, 26 (2012), 1908–75.CrossRefGoogle ScholarPubMed
Dworzak, M.N., Gaipa, G., Ratei, R., et al. Standardization of flow cytometric minimal residual disease evaluation in acute lymphoblastic leukemia, Multicentric assessment is feasible. Cytom Part B Clin Cytom, 74B (2008), 331–40.Google Scholar
Walter, R.B., Gooley, T.A., Wood, B.L., et al. Impact of pretransplantation minimal residual disease, as detected by multiparametric flow cytometry, on outcome of myeloablative hematopoietic cell transplantation for acute myeloid leukemia. J Clin Oncol, 29 (2011), 1190–7.CrossRefGoogle ScholarPubMed
Walter, R.B., Buckley, S.A., Pagel, J.M., et al. Significance of minimal residual disease before myeloablative allogeneic hematopoietic cell transplantation for AML in first and second complete remission. Blood, 122 (2013), 1813–22.Google ScholarPubMed
Walter, R.B., Gyurkocza, B., Storer, B.E., et al. Comparison of minimal residual disease as outcome predictor for AML patients in first complete remission undergoing myeloablative or nonmyeloablative allogeneic hematopoietic cell transplantation. Leukemia, 29 (2015), 137–44.CrossRefGoogle ScholarPubMed
Tobal, K., Newton, J., Macheta, M., et al. Molecular quantitation of minimal residual disease in acute myeloid leukemia with t(8,21) can identify patients in durable remission and predict clinical relapse. Blood, 95 (2000), 815–20.CrossRefGoogle Scholar
Grimwade, D., Jovanovic, J.V., Hills, R.K., et al. Prospective minimal residual disease monitoring to predict relapse of acute promyelocytic leukemia and to direct pre-emptive arsenic trioxide therapy. J Clin Oncol, 27 (2009), 3650–8.CrossRefGoogle ScholarPubMed
Buccisano, F., Maurillo, L., Principe, M.I. Del, et al. Prognostic and therapeutic implications of minimal residual disease detection in acute myeloid leukemia. Blood, 119 (2012), 332–41.CrossRefGoogle ScholarPubMed
Sievers, E.L., Lange, B.J., Alonzo, T.A., et al. Immunophenotypic evidence of leukemia after induction therapy predicts relapse, results from a prospective Children's Cancer Group study of 252 patients with acute myeloid leukemia. Blood, 101 (2003), 3398–406.CrossRefGoogle Scholar
Miyazaki, T., Fujita, H., Fujimaki, K., et al. Clinical significance of minimal residual disease detected by multidimensional flow cytometry, serial monitoring after allogeneic stem cell transplantation for acute leukemia. Leuk Res, 36 (2012), 9981003.CrossRefGoogle ScholarPubMed
Bradbury, C., Houlton, A.E., Akiki, S., et al. Prognostic value of monitoring a candidate immunophenotypic leukaemic stem/progenitor cell population in patients allografted for acute myeloid leukaemia. Leukemia, 9 (2014), 14.Google Scholar
Bastos-Oreiro, M., Perez-Corral, A., Martínez-Laperche, C., et al. Prognostic impact of minimal residual disease analysis by flow cytometry in patients with acute myeloid leukemia before and after allogeneic hemopoietic stem cell transplantation. Eur J Haematol, 93 (2014), 239–46.CrossRefGoogle ScholarPubMed
Bernal, T., Diez-Campelo, M., Godoy, V., et al. Role of minimal residual disease and chimerism after reduced-intensity and myeloablative allo-transplantation in acute myeloid leukemia and high-risk myelodysplastic syndrome. Leuk Res, 8 (2014), 551–6.Google Scholar
Ho, T-C. and Becker, M.W.. Defining patient-specific risk in acute myeloid leukemia. J Clin Oncol, 31 (2013), 3857–9.CrossRefGoogle ScholarPubMed
Lambert, J., Lambert, J., Nibourel, O., et al. MRD assessed by WT1 and NPM1 transcript levels identifies distinct outcomes in AML patients and is influenced by gemtuzumab ozogamicin. Oncotarget, 5 (2014), 6280–8.CrossRefGoogle ScholarPubMed
Hess, C.J., Feller, N., Denkers, F., et al. Correlation of minimal residual disease cell frequency with molecular genotype in patients with acute myeloid leukemia. Haematologica 94 (2009), 4653.CrossRefGoogle ScholarPubMed
Maurillo, L., Buccisano, F., Spagnoli, A., et al. Monitoring of minimal residual disease in adult acute myeloid leukemia using PB as an alternative source to BM. Haematologica, 92 (2007), 605–11.CrossRefGoogle Scholar
Gajjar, A., Ribeiro, R., Hancock, M.L., et al. Persistence of circulating blasts after 1 week of multiagent chemotherapy confers a poor prognosis in childhood acute lymphoblastic leukemia. Blood, 86 (1995), 1292–5.CrossRefGoogle ScholarPubMed
Kern, W., Haferlach, T., Schoch, C., et al. Early blast clearance by remission induction therapy is a major independent prognostic factor for both achievement of complete remission and long-term outcome in acute myeloid leukemia, data from the German AML Cooperative Group (AMLCG) 1992 Trial. Blood, 101 (2003), 6470.CrossRefGoogle Scholar
Haferlach, T., Kern, W., Schoch, C., et al. A new prognostic score for patients with acute myeloid leukemia based on cytogenetics and early blast clearance in trials of the German AML Cooperative Group. Haematologica, 89 (2003), 408–18.Google Scholar
Gianfaldoni, G., Mannelli, F., Baccini, M., et al. Clearance of leukaemic blasts from PB during standard induction treatment predicts the BM response in acute myeloid leukaemia, a pilot study. Br J Haematol, 134 (2006), 54–7.CrossRefGoogle Scholar
Gianfaldoni, G., Mannelli, F., Bencini, S., et al. PB blast clearance during induction therapy in acute myeloid leukemia. Blood, 111 (2008), 1746–7.CrossRefGoogle Scholar
Vainstein, V., Buckley, S.A., Shukron, O., et al. Rapid rate of PB blast clearance accurately predicts complete remission in acute myeloid leukemia. Leukemia, 28 (2014), 713–16.CrossRefGoogle Scholar
Lacombe, F., Arnoulet, C., Maynadié, M., et al. Early clearance of peripheral blasts measured by flow cytometry during the first week of AML induction therapy as a new independent prognostic factor, a GOELAMS study. Leukemia, 23 (2009), 350–7.CrossRefGoogle ScholarPubMed
Bertoli, S., Bories, P., Béné, M.C., et al. Prognostic impact of day 15 blast clearance in risk-adapted remission induction chemotherapy for younger patients with acute myeloid leukemia, long-term results of the multicenter prospective LAM-2001 trial by the GOELAMS study group. Haematologica, 99 (2014), 4653.CrossRefGoogle ScholarPubMed
Elliott, M.A., Litzow, M.R., Letendre, L.L., et al. Early PB blast clearance during induction chemotherapy for acute myeloid leukemia predicts superior relapse-free survival. Blood, 110 (2007), 4172–4.Google Scholar
Chou, W-C., Hou, H., Liu, C-Y., et al. Sensitive measurement of quantity dynamics of FLT3 internal tandem duplication at early time points provides prognostic information. Ann Oncol, 22 (2011), 696704.CrossRefGoogle ScholarPubMed
Krauter, J., Gorlich, K., Ottmann, O., et al. Prognostic value of minimal residual disease quantification by real-time reverse transcriptase polymerase chain reaction in patients with core binding factor leukemias. J Clin Oncol, 21 (2003), 4413–22.CrossRefGoogle ScholarPubMed
Yin, J.A.L., O'Brien, M.A., Hills, R.K., et al. Minimal residual disease monitoring by quantitative RT-PCR in core binding factor AML allows risk stratification and predicts relapse, results of the United Kingdom MRC AML-15 trial. Blood, 120 (2012), 2826–35.CrossRefGoogle ScholarPubMed
Petzer, A.L., Hogge, D.E., Landsdorp, P.M., et al. Self-renewal of primitive human hematopoietic cells (long-term-culture-initiating cells) in vitro and their expansion in defined medium. Proc Natl Acad Sci U S A, 93 (1996), 1470–4.CrossRefGoogle ScholarPubMed
Bonnet, D. and Dick, J.E.. Human acute myeloid leukemia is organized as a hierarchy that originates from a primitive hematopoietic cell. Nat Med, 3 (1997), 730–7.CrossRefGoogle ScholarPubMed
Taussig, D.C., Vargaftig, J., Miraki-Moud, F., et al. Leukemia-initiating cells from some acute myeloid leukemia patients with mutated nucleophosmin reside in the CD34(-) fraction. Blood, 115 (2010), 1976–84.CrossRefGoogle ScholarPubMed
Sarry, J-E., Murphy, K., Perry, R., et al. Human acute myelogenous leukemia stem cells are rare and heterogeneous when assayed in NOD/SCID/IL2Rγc-deficient mice. J Clin Invest, 121 (2011), 384–95.CrossRefGoogle ScholarPubMed
Terwijn, M., Zeijlemaker, W., Kelder, A., et al. Leukemic stem cell frequency, a strong biomarker for clinical outcome in acute myeloid leukemia. PLoS One, 9 (2014), e107587.CrossRefGoogle Scholar
Costello, R.T., Mallet, F., Gaugler, B., et al. Human acute myeloid leukemia CD34+/CD38- progenitor cells have decreased sensitivity to chemotherapy and Fas-induced apoptosis, reduced immunogenicity, and impaired dendritic cell transformation capacities. Cancer Res, 60 (2000), 4403–11.Google ScholarPubMed
Ishikawa, F., Yoshida, S., Saito, Y., et al. Chemotherapy-resistant human AML stem cells home to and engraft within the bone-marrow endosteal region. Nat Biotechnol, 25 (2007), 1315–21.CrossRefGoogle ScholarPubMed
Van Rhenen, A., Feller, N., Kelder, A., et al. High stem cell frequency in acute myeloid leukemia at diagnosis predicts high minimal residual disease and poor survival. Clin Cancer Res, 11 (2005), 6520–7.CrossRefGoogle ScholarPubMed
Witte, K-E., Ahlers, J., Schäfer, I., et al. High proportion of leukemic stem cells at diagnosis is correlated with unfavorable prognosis in childhood acute myeloid leukemia. Pediatr Hematol Oncol, 28 (2011), 91–9.CrossRefGoogle ScholarPubMed
Eppert, K., Takenaka, K., Lechman, E.R., et al. Stem cell gene expression programs influence clinical outcome in human leukemia. Nat Med, 17 (2011), 1086–93.CrossRefGoogle ScholarPubMed
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, 21 (2007), 1700–07.CrossRefGoogle ScholarPubMed
Bakker, A.B.H., van den Oudenrijn, S., Bakker, A.Q., et al. C-type lectin-like molecule-1, a novel myeloid cell surface marker associated with acute myeloid leukemia. Cancer Res, 64 (2004), 8443–50.Google ScholarPubMed
Larsen, H.Ø., Roug, A.S., Just, T., et al. Expression of the hMICL in acute myeloid leukemia-a highly reliable disease marker at diagnosis and during follow-up. Cytometry B Clin Cytom, 82 (2012), 38.CrossRefGoogle ScholarPubMed
Kern, W., Haferlach, T., Schnittger, S., et al. Karyotype instability between diagnosis and relapse in 117 patients with acute myeloid leukemia, implications for resistance against therapy. Leukemia, 16 (2002), 2084–91.CrossRefGoogle ScholarPubMed
Baer, M.R., Stewart, C.C., Dodge, R.K., et al. High frequency of immunophenotype changes in acute myeloid leukemia at relapse, implications for residual disease detection (Cancer and Leukemia Group B Study 8361). Blood, 97 (2001), 3574–80.CrossRefGoogle Scholar
Langebrake, C., Brinkmann, I., Teigler-Schlegel, A., et al. Immunophenotypic differences between diagnosis and relapse in childhood AML, Implications for MRD monitoring. Cytometry B Clin Cytom, 63 (2005), 19.CrossRefGoogle ScholarPubMed
Macedo, A., San Miguel, J.F., Vidriales, M-B., et al. Phenotypic changes in acute myeloid leukaemia, implications in the detection of minimal residual disease. J Clin Pathol, 49 (1996), 1518.CrossRefGoogle ScholarPubMed
Li, X., Du, W., Liu, W., et al. Comprehensive flow cytometry phenotype in acute leukemia at diagnosis and at relapse. APMIS, 118 (2010), 353–9.CrossRefGoogle ScholarPubMed
Zelezníková, T. and Babusíková, O.. The impact of cell heterogeneity and immunophenotypic changes on monitoring minimal residual disease in acute myeloid leukemia. Neoplasma, 53 (2006), 500–6.Google ScholarPubMed
Zeijlemaker, W., Gratama, J.W. and Schuurhuis, G.J.. Tumor heterogeneity makes AML a 'moving target’ for detection of residual disease. Cytometry B Clin Cytom, 86 (2014), 314.CrossRefGoogle ScholarPubMed
Oelschlägel, U., Nowak, R., Schaub, A., et al. Shift of aberrant antigen expression at relapse or at treatment failure in acute leukemia. Cytometry B Clin Cytom, 42 (2000), 247–3.3.0.CO;2-V>CrossRefGoogle ScholarPubMed
Mullighan, C.G., Phillips, L.A., Su, X., et al. Genomic analysis of the clonal origins of relapsed acute lymphoblastic leukemia. Science, 322 (2008), 1377–80.CrossRefGoogle ScholarPubMed
Ding, L., Ley, T.J., Larson, D.E., et al. Clonal evolution in relapsed acute myeloid leukemia revealed by whole genome sequencing. Nature, 481 (2012), 506–10.CrossRefGoogle ScholarPubMed
Welch, J.S., Ley, T.J., Link, D.C., et al. The origin and evolution of mutations in acute myeloid leukemia. Cell, 150 (2012), 264–78.CrossRefGoogle ScholarPubMed
Bachas, C., Schuurhuis, G.J., Assaraf, Y.G., et al. The role of minor subpopulations within the leukemic blast compartment of AML patients at initial diagnosis in the development of relapse. Leukemia, 26 (2012), 1313–20.CrossRefGoogle ScholarPubMed
Bachas, C., Schuurhuis, G.J., Hollink, I.H.I.M., et al. High-frequency type I/II mutational shifts between diagnosis and relapse are associated with outcome in pediatric AML, implications for personalized medicine. Blood, 116 (2010), 2752–8.CrossRefGoogle ScholarPubMed
Ottone, T., Zaza, S., Divona, M., et al. Identification of emerging FLT3 ITD-positive clones during clinical remission and kinetics of disease relapse in acute myeloid leukaemia with mutated nucleophosmin. Br J Haematol, 161 (2013), 533–40.CrossRefGoogle ScholarPubMed
Warren, M., Luthra, R., Yin, C.C., et al. Clinical impact of change of FLT3 mutation status in acute myeloid leukemia patients. Mod Pathol, 25 (2012), 1405–12.CrossRefGoogle ScholarPubMed
Wouters, R., Cucchi, D., Kaspers, G.J.L., et al. Relevance of leukemic stem cells in acute myeloid leukemia, heterogeneity and influence on disease monitoring, prognosis and treatment design. Expert Rev Hematol, 7 (2014), 791805.CrossRefGoogle ScholarPubMed
van Solinge, T.S., Zeijlemaker, W., Ossenkoppele, G.J., et al. The interference of genetic associations inestablishing the prognostic value of the immunophenotype in acute myeloid leukemia. Cytometry B CLin Cytom, (2017) e-pub ahead of print.Google ScholarPubMed
Ossenkoppele, G.J., van de Loosdrecht, A. and Schuurhuis, G.J.. Review of the relevance of aberrant antigen expression by flow cytometry in myeloid neoplasms. Br J Haematol, 153 (2011), 421–36.CrossRefGoogle ScholarPubMed
Lai, L., Ong, R., Li, J., et al. A CD45-based barcoding approach to multiplex mass-cytometry (CyTOF). Cytom Part A, 87 (2015), 369–74.CrossRefGoogle ScholarPubMed

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
×