A cute myeloid leukemia (AML) is a molecularly and clinically heterogeneous disease.1 Nucleophosmin (NPM1) gene mutations resulting in cytoplasmic delocalization of nucleophosmin (NPMc+)2 are the most common genetic alteration in AML, being detected in about one-third of cases. Because of its unique molecular, genotypic, immunophenotypic and prognostic features3–6 (Table 1), AML with mutated NPM1 was included as a separate provisional entity in the 2008 World Health Organization (WHO) classification of myeloid neoplasms, under the heading of “AML with recurrent genetic abnormalities”. 7 In the 2001 WHO classification, this category included only AML with t(15;17), t(8;21), inv(16), and MLL rearrangements and in 2008 it was expanded to include AMLs carrying t(6;9), inv(3) or t(3;3), AML (megakaryoblastic) with t(1;22) and two provisional entities, i.e. AML with mutated NPM1 and AML with mutated CEBPA. Thus, in the 2008 WHO classification, the category of “AML with recurrent genetic abnormalities” covers about 60% of AML.
The identification of the specific genetic alteration underlying each of the AML subtypes listed in the category of “AML with recurrent genetic abnormalities” is of critical importance since it helps to assign patients to different prognostic groups, thus influencing therapy. For example, in AML patients under 60 years of age, NPM1 mutations consistently predict favorable prognosis,6 when no concomitant FLT3-ITD mutation is present, whilst in AML patients 70 years old or over NPM1 mutations appear to be the only factor influencing prognosis in multivariate analysis.8
The search for genetic alterations using molecular methods would ideally be the gold standard for diagnosis. Unfortunately, not all centers, especially in developing countries, are equipped for molecular studies and this may hamper the worldwide use of the WHO classification. These problems could potentially be solved by developing simple surrogates for molecular studies. Here, we briefly review the molecular methods and their alternatives that are currently available for diagnosing AML with mutated NPM1.
Detection of NPM1 mutations by molecular techniques
The normal NPM1 gene configuration and the first six NPM1 mutations we identified in AML,2 which lead to common structural changes of the NPM1 protein C-terminus, are shown in Figure 1. Over the past five years, several qualitative and quantitative molecular assays for identifying NPM1 mutations have been developed and tested in a large number of AML patients.
- Qualitative assays for NPM1 mutations: these highly sensitive and specific assays for detecting NPM1 mutations9–15 are best applied to RNA or DNA extracted from fresh bone marrow or peripheral blood leukemic cells13 but plasma16 and paraffin-embedded samples17 are also suitable. More than 50 molecular variants of NPM1 mutations have been identified to date,18 mostly involving exon-12, and occasionally other exons.19 NPM1 mutations occur in about 30% of adult AML4 (50–60% of AML with normal karyotype). NPM1 mutation A (a duplication of TCTG at position 956 to 959 of the reference sequence) accounts for 75–80% of cases.2 Mutation B and D account for about 10% and 5% of cases, respectively; other mutations are very rare. NPM1 mutations are less frequent in childhood (about 8% of pediatric AML)20,21 and have been never found in children under three years of age.21 Pediatric and adult AML with mutated NPM1 appear to differ not only in frequency but also in the type of mutation as, unlike adults, the majority of children carry non-type A mutations.22 Identifying the specific type of mutation by molecular techniques is essential when PCR (Polymerase Chain Reaction)-based quantitative studies are planned (see below).
- Monitoring of minimal residual disease. Since NPM1 mutations are frequent and very stable over the course of disease,23 they are an optimal marker for monitoring minimal residual disease (MDR)24 in approximately 30% of adult AML. Indeed, the clinical value of PCR-based quantitative assessment of NPM1 mutant copies in predicting relapse and prognosis of AML with mutated NPM1 was demonstrated in several studies.25–28 Notably, Schnittger et al.28 reported the best clinical outcome was associated with the greatest reduction in the number of NPM1 mutant copies (<0.01 NPM1/ABL ratio). Monitoring of NPM1 mutant copies every 4–6 months is advisable.29
Detection of NPM1 mutated proteins by Western blot
The Western blot assay uses antibodies that recognize NPM1 mutants but not wild-type NPM1 protein in lysates from AML samples.30 These antibodies identify the specific band (37kDa) of mutated NPM1 protein only in NPM1-mutated AML cases and recognize over 95% of NPM1 mutations.30
Morphology and immunophenotype of NPM1-mutated acute myeloid leukemia
Advanced molecular or biochemical techniques are not always available or easy to apply and, therefore, a “realistic classification” based on morphological appearance was, in the past, suggested as a compromise solution for some AML subtypes.31 Since some typical FAB categories such as M3, M2 with increased eosinophils (M2eo), or M4 with increased eosinophils (M4eo), closely correlated with the presence of t(15;17), t(8;21), and inv(16), respectively, recognition of these morphological features was proposed as a surrogate for cytogenetic studies. The morphology-based approach is, however, limited by imperfect morphological-genetic correlations32 since inv(16) or t(16;16) correlate with M4eo morphology in only some cases, whilst AML with t(8;21) sometimes shows an M1 or M4 morphology. Morphology is also a poor predictor of NPM1 mutations because, although often associated with M4 and M5 morphology,2,33 AML with mutated NPM1 encompasses all other FAB categories except M3, M4eo and M7.2 Interestingly, a similar broad morphological spectrum is also seen in AML with MLL rearrangements.34
Immunophenotype combined with morphology may further increase the ability to identify specific AML genetic entities. Examples include positivity for CD19 and PAX5 in AML with t(8;21),35 low expression of HLA/DR and CD34 in acute promyelocytic leukemia (APL),7 expression of the condroitin sulfate molecule NG2 omolog (encoded by CSGP4)36 in AML with MLL rearrangements, and the consistent CD34 negativity in NPM1-mutated AML,2,4 but again the correlation immunophenotype/genotype is not complete.
Thus, in most AML cases, morphological/immunophenotypic studies alone cannot reliably predict genetic lesions, and other surrogates for molecular investigations need to be found.
Immunohistochemical detection of cytoplasmic nucleophosmin
In the 2008 WHO classification, AML with mutated NPM1 is also indicated with the synonym of NPMc+ AML (NPM cytoplasmic positive AML).7 In fact, one simple, low cost, highly-specific alternative approach to diagnosis is immunohistochemical detection of cytoplasmic nucleophosmin.37,38 This assay is fully predictive of NPM1 mutations since all molecular variants of NPM1 mutations (including those affecting exons other than 12) result in aberrant export of NPM1 mutant from the nucleus to the cytoplasm of leukemic cells.37,38 Immunohistochemistry is usually performed with monoclonal antibodies that recognize wild-type and mutated NPM1 proteins.38 Cytoplasmic nucleophosmin is optimally detected in paraffin sections from B5-fixed/EDTA-decalcified bone marrow trephines2,38,39 (Figure 2). Partial concordance between sub-cellular expression of nucleophosmin and NPM1 gene status was seen in one study40 where samples were fixed in formalin and decalcified in formic acid. It is at present unclear whether the technical problem is due to formalin fixation and/or formic acid decalcification.
Searching for cytoplasmic nucleophosmin is reminiscent of identifying APL with t(15,17) by means of the PG-M3 (anti-PML) monoclonal antibody.41 In fact, the rationale for both tests is similar. The NPM1 mutation and the PML/RAR-alpha fusion gene both cause ectopic subcellular relocalization of the respective proteins: nucleophosmin is found in cytoplasm (instead of nucleolus) and PML is observed as nuclear microspeckles (instead of nuclear bodies). In a recent study, Rego et al.42 combined morphology with the PML immunofluorescence test (PG-M3 monoclonal antibody) to investigate 102 APL patients from developing countries (Brazil, Mexico and Uruguay). Notably, this approach resulted in a more accurate and rapid diagnosis which led in turn to the immediate start of chemotherapy plus ATRA. Consequently, overall survival markedly increased from the unsatisfactory 50% in historical controls to about 80%. This study provides an example model of the big impact of using a simple test as surrogate for molecular investigations in the therapy of AML.
Immunohistochemical detection of cytoplasmic nucleophosmin may also be useful in developed countries as simple front-line screening for NPM1 mutations.37,38 The expected 30% of adults with aberrant cytoplasmic nucleophosmin could then be referred to more specialized centers for confirmation and identification of mutation type by molecular techniques. Moreover, immunohistochemistry is critical for diagnosis of AML cases presenting with “dry tap” or as myeloid sarcoma.17
As bone marrow trephines are not routinely performed in AML patients in all hematologic centers, the ability to predict NPM1 mutations from cytoplasmic expression of nucleophosmin in smears or cytospins would be particularly useful. In the present issue of Haematologica, Mattsson et al.43 found no significant correlation between sub-cellular expression of nucleophosmin and NPM1 gene status in their immunocytochemical study of smears and cytospins from 60 AMLs (31 NPM1-mutated; 29 NPM1 wild-type). Why cytoplasmic nucleophosmin is detected in fixed paraffin-embedded material but not cytological samples remains unclear. A possible explanation is that preparation and/or fixation of smear and cytospins leads to nucleophosmin diffusion across cell compartments and even out of the cells, thus preventing accurate tracking of the protein. In contrast, fixation and paraffin-embedding may optimally stabilize cell membranes (especially nuclear membrane) thus allowing accurate visualization of sub-cellular distribution of nucleophosmin.
Mattsson et al.’s43 claim that cytoplasmic nucleophosmin in paraffin sections may represent non-specific staining appears unfounded because there is much strong evidence indicating that in NPM1-mutated AML immunohistochemistry on paraffin sections depicts the real status of nucleophosmin sub-cellular distribution. In fact, aberrant cytoplasmic expression of nucleophosmin in B5 fixed/EDTA decalcified bone marrow trephines is fully predictive of NPM1 mutations.37,39 Aberrant nuclear export of nucleophosmin is consistent with the molecular alterations at the C-terminus of all NPM1 mutant proteins (Figure 1) including those generated by NPM1 mutations involving exons other than exon 12.38 Moreover, antibodies that specifically recognize mutant but not wild-type NPM1 protein, consistently label the cytoplasm of NPM1-mutated AML cells at both immunohistochemistry44 and Western blotting.30 Finally, laser confocal microscopy of transfected cells clearly showed that fluorescent-tagged wild-type and mutated NPM1 proteins localized respectively in the nucleolar and cytoplasmic cell compartments. 4,38
Whatever the reason for the failure to detect cytoplasmic nucleophosmin in smears or cytospins, the comprehensive study by Mattsson et al.43 clearly indicates this method cannot be at present recommended as surrogate marker for NPM1 mutations. One promising approach based on the use of intracellular flow cytometry for rapid, specific detection of nucleophosmin in the cytoplasm of leukemic cells has been recently proposed as an alternative to bone marrow trephines in the diagnosis of NPM1-mutated AML.45 Because of its simplicity, this test may emerge in the future as a valuable surrogate to molecular studies for initial screening of AML samples. Another method that can be used in alternative to bone marrow trephines for detection of cytoplasmic nucleophosmin is immunostaining of sections cut from paraffin-embedded pellets of peripheral blood leukemic cells (B. Falini, unpublished observation, 2009).
Surrogates for molecular studies are expected to be particularly useful in older patients where NPM1 mutations appear to play a prognostic role independently of the FLT3 gene status.8 For younger patients, whose favorable prognosis is associated with the NPM1-mutated/FLT3-ITD negative genotype,4,6 we must search for rapid, inexpensive assays which can also allow us to assess the FLT3 gene status, which is still only evaluable by molecular methods.
Conclusions
Because of its distinctive features, AML with mutated NPM1 (NPMc+ AML) was included as a provisional entity in the 2008 WHO classification of myeloid neoplasms. Over the past five years several methods have been developed to diagnose this new entity. NPM1 mutations can be detected by molecular techniques or surrogates such as immunohistochemistry, Western blotting and possibly flow cytometry. These methods are complementary rather than competitive and offer a flexible approach to diagnosis which is essential if the WHO Classification is to be implemented, as intended, worldwide.
Footnotes
- Brunangelo Falini is Professor of Hematology at the University of Perugia, Perugia, Italy. Maria Paola Martelli is a Researcher in Hematology at the University of Perugia, Perugia, Italy. Stefano A Pileri is Professor of Pathology and Director of the Hematopathology Unit at Bologna University, Bologna, Italy. Cristina Mecucci is Associated Professor of Hematology at the University of Perugia, Perugia, Italy.
- Drs. Falini and Mecucci applied for a patent on the clinical use of NPM1 mutants. No other potential conflicts of interests relevant to this article were reported.
- Supported by the Associazione Italiana per la Ricerca sul Cancro (A.I.R.C.) and Fondazione Cassa di Risparmio di Perugia (Grants n. 2007.0099.020 and 2008.020.058). The authors would like to thank Claudia Tibido’ for her excellent secretarial assistance and Dr. Geraldine Anne Boyd for her help in editing this paper. The authors apologize to those whose papers could not be cited owing to space limitations.
- ( Related Original Article on page 670)
References
- Lowenberg B. Acute myeloid leukemia: the challenge of capturing disease variety. Hematology Am Soc Hematol Educ Program. 2008:1-11. Google Scholar
- Falini B, Mecucci C, Tiacci E, Alcalay M, Rosati R, Pasqualucci L. Cytoplasmic nucleophosmin in acute myelogenous leukemia with a normal karyotype. N Engl J Med. 2005; 352(3):254-66. PubMedhttps://doi.org/10.1056/NEJMoa041974Google Scholar
- Alcalay M, Tiacci E, Bergomas R, Bigerna B, Venturini E, Minardi SP. 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(3):899-902. PubMedhttps://doi.org/10.1182/blood-2005-02-0560Google Scholar
- Falini B, Nicoletti I, Martelli MF, Mecucci C. Acute myeloid leukemia carrying cytoplasmic/mutated nucleophosmin (NPMc+ AML): biologic and clinical features. Blood. 2007; 109(3):874-85. PubMedhttps://doi.org/10.1182/blood-2006-07-012252Google Scholar
- Garzon R, Garofalo M, Martelli MP, Briesewitz R, Wang L, Fernandez-Cymering C. Distinctive microRNA signature of acute myeloid leukemia bearing cytoplasmic mutated nucleophosmin. Proc Natl Acad Sci USA. 2008; 105(10):3945-50. PubMedhttps://doi.org/10.1073/pnas.0800135105Google Scholar
- Schlenk RF, Dohner K, Krauter J, Frohling S, Corbacioglu A, Bullinger L. Mutations and treatment outcome in cytogenetically normal acute myeloid leukemia. N Engl J Med. 2008; 358(18):1909-18. PubMedhttps://doi.org/10.1056/NEJMoa074306Google Scholar
- WHO Classification of Tumours of Haematopoietic and Lymphoid Tissues. International Agency for Research on Cancer (IARC): Lyon; 2008. Google Scholar
- Becker H, Marcucci G, Maharry K, Radmacher MD, Mrozek K, Margeson D. Favorable prognostic impact of NPM1 mutations in older patients with cytogenetically normal de novo acute myeloid leukemia and associated gene- and microRNA-expression signatures: a cancer and leukemia group B study. J Clin Oncol. 2010; 28(4):596-604. PubMedhttps://doi.org/10.1200/JCO.2009.25.1496Google Scholar
- Roti G, Rosati R, Bonasso R, Gorello P, Diverio D, Martelli MF. Denaturing high-performance liquid chromatography: a valid approach for identifying NPM1 mutations in acute myeloid leukemia. J Mol Diagn. 2006; 8(2):254-9. PubMedhttps://doi.org/10.2353/jmoldx.2006.050098Google Scholar
- Laughlin TS, Becker MW, Liesveld JL, Mulford DA, Abboud CN, Brown P. Rapid method for detection of mutations in the nucleophosmin gene in acute myeloid leukemia. J Mol Diagn. 2008; 10(4):338-45. PubMedhttps://doi.org/10.2353/jmoldx.2008.070175Google Scholar
- Ottone T, Ammatuna E, Lavorgna S, Noguera NI, Buccisano F, Venditti A. An allele-specific rt-PCR assay to detect type A mutation of the nucleophosmin-1 gene in acute myeloid leukemia. J Mol Diagn. 2008; 10(3):212-6. PubMedhttps://doi.org/10.2353/jmoldx.2008.070166Google Scholar
- Szankasi P, Jama M, Bahler DW. A new DNA-based test for detection of nucleophosmin exon 12 mutations by capillary electrophoresis. J Mol Diagn. 2008; 10(3):236-41. PubMedhttps://doi.org/10.2353/jmoldx.2008.070167Google Scholar
- Wertheim G, Bagg A. Nucleophosmin (NPM1) mutations in acute myeloid leukemia: an ongoing (cytoplasmic) tale of dueling mutations and duality of molecular genetic testing methodologies. J Mol Diagn. 2008; 10(3):198-202. PubMedhttps://doi.org/10.2353/jmoldx.2008.080019Google Scholar
- Huang Q, Chen W, Gaal KK, Slovak ML, Stein A, Weiss LM. A rapid, one step assay for simultaneous detection of FLT3/ITD and NPM1 mutations in AML with normal cytogenetics. Br J Haematol. 2008; 142(3):489-92. PubMedhttps://doi.org/10.1111/j.1365-2141.2008.07205.xGoogle Scholar
- Dvorakova D, Lengerova M, Pospisilova J, Palasek I, Mayer J. A novel quantitative assessment of minimal residual disease in patients with acute myeloid leukemia carrying NPM1 (nucleophosmin) exon 12 mutations. Leukemia. 2009; 23(4):793-6. PubMedhttps://doi.org/10.1038/leu.2008.268Google Scholar
- Ma W, Kantarjian H, Zhang X, Jilani I, Sheikholeslami MR, Donahue AC. Detection of nucleophosmin gene mutations in plasma from patients with acute myeloid leukemia: clinical significance and implications. Cancer Biomark. 2009; 5(1):51-8. PubMedGoogle Scholar
- Falini B, Lenze D, Hasserjian R, Coupland S, Jaehne D, Soupir C. Cytoplasmic mutated nucleophosmin (NPM) defines the molecular status of a significant fraction of myeloid sarcomas. Leukemia. 2007; 21(7):1566-70. PubMedhttps://doi.org/10.1038/sj.leu.2404699Google Scholar
- Rau R, Brown P. Nucleophosmin (NPM1) mutations in adult and childhood acute myeloid leukaemia: towards definition of a new leukaemia entity. Hematol Oncol. 2009; 27(4):171-81. PubMedhttps://doi.org/10.1002/hon.904Google Scholar
- Albiero E, Madeo D, Bolli N, Giaretta I, Bona ED, Martelli MF. Identification and functional characterization of a cytoplasmic nucleophosmin leukaemic mutant generated by a novel exon-11 NPM1 mutation. Leukemia. 2007; 21(5):1099-103. PubMedGoogle Scholar
- Cazzaniga G, Dell'Oro MG, Mecucci C, Giarin E, Masetti R, Rossi V. Nucleophosmin mutations in childhood acute myelogenous leukemia with normal karyotype. Blood. 2005; 106(4):1419-22. PubMedhttps://doi.org/10.1182/blood-2005-03-0899Google Scholar
- Brown P, McIntyre E, Rau R, Meshinchi S, Lacayo N, Dahl G. The incidence and clinical significance of nucleophosmin mutations in childhood AML. Blood. 2007; 110(3):979-85. PubMedhttps://doi.org/10.1182/blood-2007-02-076604Google Scholar
- Thiede C, Creutzig E, Reinhardt D, Ehninger G, Creutzig U. Different types of NPM1 mutations in children and adults: evidence for an effect of patient age on the prevalence of the TCTG-tandem duplication in NPM1-exon 12. Leukemia. 2007; 21(2):366-7. PubMedhttps://doi.org/10.1038/sj.leu.2404519Google Scholar
- Falini B, Martelli MP, Mecucci C, Liso A, Bolli N, Bigerna B. Cytoplasmic mutated nucleophosmin is stable in primary leukemic cells and in a xenotransplant model of NPMc+ acute myeloid leukemia in SCID mice. Haematologica. 2008; 93(5):775-9. PubMedhttps://doi.org/10.3324/haematol.12225Google Scholar
- Gorello P, Cazzaniga G, Alberti F, Dell’Oro MG, Gottardi E, Specchia G. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations. Leukemia. 2006; 20(6):1103-8. PubMedhttps://doi.org/10.1038/sj.leu.2404149Google Scholar
- Chou WC, Tang JL, Wu SJ, Tsay W, Yao M, Huang SY. Clinical implications of minimal residual disease monitoring by quantitative polymerase chain reaction in acute myeloid leukemia patients bearing nucleophosmin (NPM1) mutations. Leukemia. 2007; 21(5):998-1004. PubMedGoogle Scholar
- Barragan E, Pajuelo JC, Ballester S, Fuster O, Cervera J, Moscardo F. Minimal residual disease detection in acute myeloid leukemia by mutant nucleophosmin (NPM1): comparison with WT1 gene expression. Clin Chim Acta. 2008; 395(1–2):120-3. PubMedhttps://doi.org/10.1016/j.cca.2008.05.021Google Scholar
- Bacher U, Badbaran A, Fehse B, Zabelina T, Zander AR, Kroger N. Quantitative monitoring of NPM1 mutations provides a valid minimal residual disease parameter following allogeneic stem cell transplantation. Exp Hematol. 2009; 37(1):135-42. PubMedhttps://doi.org/10.1016/j.exphem.2008.09.014Google Scholar
- Schnittger S, Kern W, Tschulik C, Weiss T, Dicker F, Falini B. Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML. Blood. 2009; 114(11):2220-31. PubMedhttps://doi.org/10.1182/blood-2009-03-213389Google Scholar
- Ommen HB, Schnittger S, Jovanovic JV, Ommen IB, Hasle H, Ostergaard M. Strikingly different molecular relapse kinetics in NPM1c, PML-RARA, RUNX1-RUNX1T1 and CBFB-MYH11 acute myeloid leukemias. Blood. 2010; 115(2):198-205. PubMedhttps://doi.org/10.1182/blood-2009-04-212530Google Scholar
- Martelli MP, Manes N, Liso A, Pettirossi V, Verducci Galletti B, Bigerna B. A western blot assay for detecting mutant nucleophosmin (NPM1) proteins in acute myeloid leukaemia. Leukemia. 2008; 22(12):2285-8. PubMedhttps://doi.org/10.1038/leu.2008.149Google Scholar
- Arber DA. Realistic pathologic classification of acute myeloid leukemias. Am J Clin Pathol. 2001; 115(4):552-60. PubMedhttps://doi.org/10.1309/K2PH-L2U7-722B-B1YRGoogle Scholar
- Vardiman JW, Harris NL, Brunning RD. The World Health Organization (WHO) classification of the myeloid neoplasms. Blood. 2002; 100(7):2292-302. PubMedhttps://doi.org/10.1182/blood-2002-04-1199Google Scholar
- Schnittger S, Schoch C, Kern W, Mecucci C, Tschulik C, Martelli MF. Nucleophosmin gene mutations are predictors of favorable prognosis in acute myelogenous leukemia with a normal karyotype. Blood. 2005; 106(12):3733-9. PubMedhttps://doi.org/10.1182/blood-2005-06-2248Google Scholar
- Schoch C, Schnittger S, Kern W, Dugas M, Hiddemann W, Haferlach T. Acute myeloid leukemia with recurring chromosome abnormalities as defined by the WHO-classification: incidence of subgroups, additional genetic abnormalities, FAB subtypes and age distribution in an unselected series of 1,897 patients with acute myeloid leukemia. Haematologica. 2003; 88(3):351-2. PubMedGoogle Scholar
- Tiacci E, Pileri S, Orleth A, Pacini R, Tabarrini A, Frenguelli F. PAX5 expression in acute leukemias: higher B-lineage specificity than CD79a and selective association with t(8;21)-acute myelogenous leukemia. Cancer Res. 2004; 64(20):7399-404. PubMedhttps://doi.org/10.1158/0008-5472.CAN-04-1865Google Scholar
- Wuchter C, Harbott J, Schoch C, Schnittger S, Borkhardt A, Karawajew L. Detection of acute leukemia cells with mixed lineage leukemia (MLL) gene rearrangements by flow cytometry using monoclonal antibody 7.1. Leukemia. 2000; 14(7):1232-8. PubMedhttps://doi.org/10.1038/sj.leu.2401840Google Scholar
- Falini B, Martelli MP, Bolli N, Bonasso R, Ghia E, Pallotta MT. Immunohistochemistry predicts nucleophosmin (NPM) mutations in acute myeloid leukemia. Blood. 2006; 108(6):1999-2005. PubMedhttps://doi.org/10.1182/blood-2006-03-007013Google Scholar
- Falini B, Bolli N, Liso A, Martelli MP, Mannucci R, Pileri S. Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1: molecular basis and clinical implications. Leukemia. 2009; 23(10):1731-43. PubMedhttps://doi.org/10.1038/leu.2009.124Google Scholar
- Luo J, Qi C, Xu W, Kamel-Reid S, Brandwein J, Chang H. Cytoplasmic expression of nucleophosmin accurately predicts mutation in the nucleophosmin gene in patients with acute myeloid leukemia and normal karyotype. Am J Clin Pathol. 2010; 133(1):34-40. PubMedhttps://doi.org/10.1309/AJCPCI1FFE2DRXIVGoogle Scholar
- Konoplev S, Huang X, Drabkin HA, Koeppen H, Jones D, Kantarjian HM. Cytoplasmic localization of nucleophosmin in bone marrow blasts of acute myeloid leukemia patients is not completely concordant with NPM1 mutation and is not predictive of prognosis. Cancer. 2009; 115(20):4737-44. PubMedhttps://doi.org/10.1002/cncr.24543Google Scholar
- Falini B, Flenghi L, Fagioli M, Lo Coco F, Cordone I, Diverio D. Immunocytochemical diagnosis of acute promyelocytic leukemia (M3) with the monoclonal antibody PG-M3 (anti-PML). Blood. 1997; 90(10):4046-53. PubMedGoogle Scholar
- Rego EM, Kim HT, Ruiz-Arguelles GJ, Uriarte R, Jacomo RH, Gutierrez-Aguirre H. Improving the treatment outcome of acute promyelocytic leukemia in developing countries through International Cooperative Network. Report on the International Consortium on Acute Promyelocytic Leukemia Study Group. Blood (ASH Annual Meeting Abstracts). 2009; 114:6. Google Scholar
- Mattsson G, Turner SH, Cordell J, Ferguson DJ, Schuh A, Grimwade LF. Can cytoplasmic nucleophosmin be detected by immunocytochemical staining of cell smears in acute myeloid leukemia?. Haematologica. 2010; 95(4):670-3. PubMedhttps://doi.org/10.3324/haematol.2009.011817Google Scholar
- Pasqualucci L, Liso A, Martelli MP, Bolli N, Pacini R, Tabarrini A. Mutated nucleophosmin detects clonal multilineage involvement in acute myeloid leukemia: Impact on WHO classification. Blood. 2006; 108(13):4146-55. PubMedhttps://doi.org/10.1182/blood-2006-06-026716Google Scholar
- Oelschlaegel U KS, Schaich M, Kroschinsky F, Parmentier S, Ehninger G, Thiede C. A rapid flow cytometric method for the detection of NPM1 mutated patients with acute myeloid leukemia (AML). Blood (ASH Annual Meeting Abstracts). 2008; 112:1490. Google Scholar