NPM1 Mutated, BCR-ABL1 Positive Myeloid Neoplasms: Review of the Literature 

Gianfranco Catalano1,2,3, Pasquale Niscola3, Cristina Banella1,2, Daniela Diverio4, Malgorzata Monika Trawinska, Stefano Fratoni5, Rita Iazzoni6, Paolo De Fabritiis1,3, Elisabetta Abruzzese3* and Nelida Ines Noguera1,2*.

1 Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00133 Rome, Italy.
2 Neuro Oncohematology Unit, Santa Lucia Foundation, IRCCS. Rome, Italy.
3 Hematology Unit, Sant’ Eugenio Hospital, Tor Vergata University of Rome, Rome, Italy.
4 Hematology, Department of Precision and Translational Medicine, Policlinico Umberto I, “Sapienza” University of Rome, Rome, Italy.
5 Department of Pathology (UOSD Anatomia Patologica) A.S.L. Roma2, Sant’ Eugenio Hospital, Rome, Italy.
6 Department of Clinical Pathology (U.O.C. Laboratorio) A.S.L. Roma2, Sant’ Eugenio Hospital, Rome, Italy.   

Correspondence to: Nelida Ines Noguera. Dept. of Biomedicine and Prevention, Tor Vergata University, 00133 Rome, Italy. Tel: +3906501703214, Fax: +3906501703318. E-mail: n.noguera@hsantalucia.it.
Elisabetta Abruzzese. Hematology Unit, Saint’ Eugenio Hospital, Tor Vergata University of Rome, 00133 Rome, Italy. E-mail: elisabetta.abruzzese@uniroma2.it  

Published: November 1, 2020
Received: August 26, 2020
Accepted: October 22, 2020
Mediterr J Hematol Infect Dis 2020, 12(1): e2020083 DOI 10.4084/MJHID.2020.083

This is an Open Access article distributed under the terms of the Creative Commons Attribution License
(
https://creativecommons.org/licenses/by-nc/4.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Abstract

Breakpoint cluster region - Abelson (BCR-ABL1) chimeric protein and mutated Nucleophosmin (NPM1) are often present in hematological cancers, but they rarely coexist in the same disease. Both anomalies are considered founder mutations that inhibit differentiation and apoptosis, but BCR-ABL1 could act as a secondary mutation conferring a proliferative advantage to a pre-neoplastic clone. The 2016 World Health Organization (WHO) classification lists the provisional acute myeloid leukemia (AML) with BCR-ABL1, which must be diagnosed differentially from the rare blast phase (BP) onset of chronic myeloid leukemia (CML), mainly because of the different therapeutic approach in the use of tyrosine kinase inhibitors (TKI). Here we review the BCR/ABL1 plus NPMc+ published cases since 1975 and describe a case from our institution in order to discuss the clinical and molecular features of this rare combination, and report the latest acquisition about an occurrence that could pertain either to the rare AML BCR-ABL1 positive or the even rarer CML-BP with mutated NPM1 at the onset. Differential diagnosis is based on careful analysis of genotypic and phenotypic features and anamnestic, clinical evolution, and background data. Therapeutic decisions must consider the broader clinical aspects, the comparatively mild effects of TKI therapy versus the great benefit that might bring to most of the patients, as may be incidentally demonstrated by our case history.



Introduction

Mutations in the NPM1 gene are the most frequent genetic abnormalities in acute myeloid leukemia (AML) and are highly specific for de novo AML.[1] The Breakpoint cluster region - Abelson (BCR-ABL) fusion gene is the genetic hallmark of chronic myeloid leukemia (CML) but can also be found in approximately 30% of acute lymphoblastic leukemia (ALL) and rarely in AML (0.3–3% of newly diagnosed cases).[2–4] In the updated World Health Organization (WHO) classification published in 2016, AML with BCR-ABL has been introduced as a provisional new entity.[5,6] To the best of our knowledge, the co-occurrence of the BCR-ABL fusion gene and NPM1 mutations in de novo AML has been reported in only a few cases. In this review, we analyzed all the BCR/ABL1 plus NPMc+ published cases since 1975 and a case from our institution to present common clinical and molecular features of this rare disease.
t(9;22)(q34.1;q11.2) BCR-ABL. Among human cancers, AMLs are relatively genetically simple and stable diseases featuring the fewest mutations variety and average. AML genomes contain a median of 13 coding mutations (single nucleotide variants and insertion/deletions) and an average of less than one gene-fusion event.[7,8] Most of the fusions derive from translocation events, and Philadelphia chromosome t(9;22)(q34.1;q11.2), generating the BCR-ABL1 chimeric protein, was the first genetic aberration associated with human cancer: Chronic myeloid leukemia (CML). BCR-ABL activates proliferation signaling pathways (RAS and STAT5, STAT1 and STAT6 signaling, PI3-K and AKT/PKB pathways), analogously to PML/RARa in APL inhibits PTEN,[9,10] interferes with the focal adhesion complex (PAXILLIN, FAK), induces abnormal integrin signaling (FAK/CRK-L/SDF-1) and has anti-apoptotic activity (PI63K/Akt/STAT5). In addition, BCR-ABL has been shown to generate a “mutator” phenotype downregulating homeostatic controls and DNA repair pathways and promoting the expression of DNA-polymerase-beta, which is prone to copy errors during DNA replication.[11] Since no CML-BP with lymphoid phenotype carrying the NPM1c+ mutation was ever reported, we will not address the subject of Ph1+ALL. Contrarily to what could be expected only a few years ago, myeloid neoplasms carrying the BCR-ABL transcripts are a composite subset of hematological disorders. If the principal disease, CML, in which BCR-ABL1 is involved, has well-characterized features and standardized diagnosis and therapy, the picture is composite for the other neoplastic diseases. 2016 WHO classification of myeloid neoplasms and acute leukemia distinguish, other than CML, two more entities: one mixed phenotype acute leukemia (MPAL) with BCR-ABL1, and the provisional AML with BCR-ABL1.[6] Since no CML-BP with lymphoid phenotype carrying the NPM1c+ mutation was ever reported, we will not address the subject of Ph1+ALL.
Nucleophosmin. Nucleophosmin (NPM1) is present in high quantities in the granular region of nucleoli but shuttles between nucleus and cytoplasm, acting as a chaperone. Chaperones are molecules that associate with target proteins, organize their structure, convoy them to the appropriate place, and molecular aggregate but are not part and have no function in that aggregate.[12,13] NPM1 has been identified as the most frequently mutated gene in AML patients, accounting for about 30% of cases,[14–16] the vast majority of which with normal karyotype. At onset, NPM1 mutation associates with a less severe prognosis, but clonal evolution can lead to additional genetic abnormalities and worst prognosis.[1,16,17] NPM1 gene mutations in AML lead to a new C-terminus sequence in the mutant protein, that, as compared to the wild-type protein, lacks the nucleolar binding site and acquires a nuclear export signal: mutated NPM1 is confined to cytoplasm, its absence from the nucleus seems to be the basis for the oncogenic phenotype since the protein plays a role in chromatin remodeling, centrosome duplication, DNA replication, recombination, transcription, and repair as well as in the control of cell cycle progression and survival in response to a variety of stress stimuli.[12,18–21]
The Paradigm of Leukemogenesis. Mutations within a cell can influence the rate of acquisition of other lesions. After the initiating mutation, there might be a gradual accumulation of additional genetic alterations or accelerated progression due to genomic instability or catastrophic genetic events, including chromothripsis.[22–27] The number of identifiable driver mutations differs between AML cases. Although most cases harbor three or more identifiable drivers at the time of clinical presentation, human sequencing data describe many AML with only one or two identifiable driver mutations.[24,28] According to the model of Gilliland and Griffin, the paradigm of leukemogenesis features a class II mutation as leukemia-initiating event, causing inhibition of differentiation and apoptosis, cooperating with a class I mutations conferring a proliferative advantage to the clone.[29,30] In 2013 the Cancer Genome Atlas Research Network classified three sets of genes with the strongest patterns of mutual exclusivity. For the purpose, they used whole genome or whole exome sequencing and statistical analysis of 200 de novo AML cases selected from a set of more than 400 samples to reflect a real-world distribution of subtypes. The first set comprised the transcription-factor fusion genes and mutations involving NPM1, RUNX1, TP53, and CEBPA, the second set the mutations in genes encoding FLT3 or other tyrosine kinases (TK), serine-threonine kinases, protein tyrosine phosphatases, RAS family proteins, and the third set included mutations in ASXL1 and genes encoding components of the cohesin complex, other myeloid transcription factors, and other epigenetic modifiers.[7] The association of BCR-ABL1 and mutated NPM1 in the same clone is unusual but not contradictory to either of the models if NPM1 is the founder, class II mutation, and BCR-ABL1 acts a class I mutation, conferring a proliferative advantage to the affected cells. BCR-ABL1, even though capable of transforming hemopoietic stem cells single-handed and causing per se CML and diverse acute leukemias (Ph1+ ALL, MPAL and AML), could be working as a class I mutation[31] since the molecular aberration was found in tumor subclones and even in oligoclones in otherwise normal bone marrow.[32,33] However, would that be possible to reverse the rank of the mutations, as in a CML blastic phase (CML-BP) clone carrying mutated NPM1 evolving from an NPM1-negative chronic phase disease? Moreover, how to discriminate between de novo Philadelphia positive AML and a CML diagnosed at BP onset? Even though identical regarding two substantial features of the genetic profile, the two conditions must have different biology. The presence of BCR-ABL1 protein ab initio, thus in tumor-initiating cells and all the disease clones, must confer the phenotype, natural history, and clinic of CML. Conversely, the emergence of a BCR-ABL positive clone as a type I mutation in an NPM1 mutation expressing clone is not more than a concomitant feature in the characteristic of acute leukemia, in a way not entirely different from a FLT3 activating mutation (Figure 1).

Figure 1 Figure 1.  Clonal evolution patterns for AML and CML.
A- BCR-ABL1 causes a “mutator” phenotype downregulating homeostatic controls and DNA repair pathways. Several subclones develop until additional mutations (double Ph1, +8, +19, +21, i(17q), abnormalities of chromosome 7, mutation of TP53, RB1, MYC, CDKN2A, RAS, RUNX1, and EVI1 genes) generate the blastic phase clone. The disease is often chemoresistant, and TKI therapy does not always work, but is still the best therapeutic option, particularly in TKI naïve patients. After remission, the disease could be controlled for a lasting remission; emerging resistant clones can be controlled with a second TKI (*) or develop into a full relapse of the BP(**). B- The patient, here described, responded to induction therapy and maintained continuous remission under TKI therapy. C- Major clonal evolution patterns during AML insurgence and relapse implicate that the disease’s founding clone gains mutations and evolves into the disease. After remission, a relapse clone(s) could arise from the original under the selective pressure of therapy. Otherwise, a subclone of the initial clone survives therapy, gains additional mutations, and expands at relapse. In both models, a founding type II mutation is followed by cooperating type I mutations able to convey a proliferative advantage. Often in BRC-ABL1+ AML patients, the relapsing clone lacks BCR-ABL1 mutation and therefore is insensitive to TKI therapy.


NPM1 Mutated and BCR-ABL1 Positive Myeloid Neoplasm

To the best of our knowledge, only a few cases of de novo AML with BCR-ABL1 and NPM1 mutations were published in the last decades (Table 1). Bacher et al. describe a case with normal karyotype AML (FAB M4) and an NPM1 mutation, the occurrence of a Philadelphia positive subclone in an NPM1 mutated AML patient emerging at relapse of the disease. This patient received initially high dose of chemotherapy, and also intensive chemotherapy with high dose cytarabine and mitoxantrone after relapse, unfortunately dying from progression 26 months from diagnosis.[31] Single cases with Philadelphia positive subclones in NPM1 mutated AML had previously been reported by Suzuki et al.[34] and Verhaak et al. [35] Palmisano et al. reported a patient who maintained the NPM1 mutation at relapse of the disease, whereas the t(9;22) was lost.[36] Konoplev et al. analyzed NPM1 and ABL1 genes, often mutated in AML and CML-BP patients, respectively, to gather insights into the relationship between Ph+ AML and CML-BP. They studied 9 Ph+ AML and 5 CML-BP patients at the onset. Two out of 9 Ph+ AML patients had NPM1 mutations and were alive 36 and 71 months after diagnosis. All Ph+ AML had no mutation in the ABL1 sequence, no NPM1 mutations were identified in the CML-BP group, and one CML-BP patient had ABL1 mutation. The Authors argue that Ph+ AML is distinct from CML-BP.[36] Reboursiere et al. describe one case harboring a BCR-ABL p210 transcript level of approximately 10% with NPM1 gene mutation. The patient received induction therapy with mitoxantrone, daunorubicin, and cytosine arabinoside and two courses of high-dose cytosine arabinoside consolidation therapy followed by allogenic hematopoietic stem cell transplantation (allo-HSCT). Eleven years after allo-HSCT, the patient remained in continuous complete molecular remission.[37] Mattioti et al. report a patient diagnosed with AML harboring a complex three-way translocation t(9;22;12)(q34;q13;q11) encoding for two isoforms of BCR-ABL transcript (b3a2;b2a2) and a concomitant type A mutation in the NPM1 gene. The patient was started on initial cytoreductive treatment with hydroxyurea for ten days and was subsequently treated with second-generation tyrosine kinase inhibitor (TKI) dasatinib due to the central nervous system’s high risk involvement and extramedullary localization. Unfortunately, the patient died after 3 months of treatment.[38] Studies regarding CML have shown that in some cases transcript, b2a2 has slower molecular and inferior response rates to TKI and a poorer long-term outcome,[39] but at present, no reliable data are available regarding the prognostic value of the different transcripts in AML. Neuendorff et al., in an exhaustive review, describe 6 NPM1 mutated at primary diagnosis out of 126 cases of de novo BCR-ABL1+ AML. At least 3 of these 6 NPM1 BCR-ABL1+ AML patients were long-term survivors, which notwithstanding the exiguity of cases, is a large percentage.[40]

Table 1 Table 1.AML with BCR-ABL1 and NPM1 mutations.

From the clinical point of view, sometimes the presence of the BCR-ABL1 hybrid transcript could not clarify the adjudication since a precise distinction between AML and CML-BP at onset is still to be defined, which poses problems of diagnosis and therapy, most of all about the timing and efficacy of TKI therapy. Notwithstanding the rarity of cases, it seems clear that AML with BCR-ABL1, in general, does not always respond well to TKI therapy ;[40] conversely, a TKI naïve CML must benefit from TKI therapy.[41]
Experience in Our Institution. Here we want to narrate the case of a patient, 43 years of age, male, diagnosed in April 2013 at a different country institution supposedly with a CML onset in BP and treated with 3 days Idarubicin plus seven days of high dose Aracytin (IA 3+7) resulting in complete hematological remission on day 26. The patient had experienced a series of severe complications during the induction therapy. The routine molecular assessment had documented positivity for BCR-ABL1 (p210-B3A2) translocation and NPM1 mutation A.[42,43] The patient, with residual hepatic toxicity, was prescribed Dasatinib 100mgr as maintenance therapy. The diagnosis was well documented as for the extension of the search for the mutations but essential since we were not detailed about the FAB and immunophenotype of the blasts, quantities of the mutated transcripts, or about any other clinical aspect that would explain the choice of CML-BP over AML with BCR-ABL1+. In our opinion is of interest that the case was labeled as CML-BP, yet the clinical perspective and indication were that of a de novo AML with BCR-ABL1: intensive induction chemotherapy with additional TKI maintenance therapy, then consolidation and allo-SCT.
On day 45, our bone marrow evaluation showed hematologic morphologic remission, molecular remission of the BCR-ABL1 transcript (p210 transcript was 0.069% with MR3 sensitivity, p190 was 0,0% negative MR3 sensitivity) and molecular remission of the NPM1 mutated transcript (0.025 of NPM1 mutation A copies every 104 copies of ABL, cut off value 0.03).[44] Of note that only a small percentage, less than 10%, of CML-BP patients, achieves molecular remission after frontline chemotherapy plus TKI therapy.[41] In contrast, almost one in three of AML NPM1 mutated patients achieve molecular remission 30 days after frontline therapy.[45] The search for a compatible donor among siblings was unsuccessful, we consulted with the patient. We agreed to postpone intensive chemotherapy mainly for the high risk of a recurrence of the intestinal bleeding, at the same time the patient did not agree to start a search for an unrelated compatible donor. One year since onset p210 transcript was undetectable with MR5 sensitivity, we stop essaying p190, and the NPM1 mutated transcript remained in molecular remission (0.03 of NPM1 mutation A copies every 104 copies of ABL, cut off value 0.03). After more than 6 years, the patient is still in continuous profound molecular remission of the BCR-ABL1 (undetectable transcript with MR5 sensitivity) and remission of the NPM1 transcripts. Still assuming Dasatinib, the dose was interrupted for 30 days and then reduced by half to 50 mg per day due to a chemical pleuritis about 12 months ago. The interruption and lowered dose did not cause any variation in the molecular remission of both transcripts. Since all disease-free survival (DFS) curves tend to plateau after 2-3 years of follow-up and relapse after 5 years of DFS are rare events, we may say the patient was fortunate not to undergo intensive chemotherapy and allo-SCT.
Nevertheless, is this patient eligible for ending TKI therapy? It all depends on the diagnosis. A CML-BP in remission is not eligible in any case for stopping TKI therapy. Conversely, an AML in continuous molecular remission after 6 years could be considered for ending the treatment.

Differential Diagnosis

CML primary blast phase is an infrequent event, and the secondary blast phase is usually marked out by patients’ medical history. In the lack of a previous CML diagnosis, most BP cases must carry the clinical and morphologic stigmata of the chronic phase. Thus anamnestic and clinical features, histology and morphology, immunophenotype, and genetic can be of help. The presence of basophilia often accompanies blast cell expansion and disease acceleration in CML, whereas it is not a common feature in de novo AML, the same for splenomegaly. In BP, bone marrow megakaryocytic count is increased in most cases with perisinusoidal distribution and no clusters, hypolobation of nuclei, and the presence of micromegakaryocytes. Broadly 75% of BP show a myeloid phenotype and in more than 80% of cases feature additional genetic abnormalities (double Ph1, +8, +19, +21, i(17q), abnormalities of chromosome 7, mutation of TP53, RB1, MYC, CDKN2A, RAS, RUNX1, and EVI1 genes).[46] Of note, ABL1 TK domain mutations are typical of CML-BP and are not restricted to patients with prior TKI exposure. In a study of unselected TKI-naïve CML-BP patients, 5 of 19 patients had ABL1 mutations.[45] ABL1 mutations have not been reported in Ph+ AML patients.[15] Under the genotypic profile, there is a link between BCR-ABL1 rearrangement and some features associated with the lymphoid phenotype. Nacheva et al. studying 9 de novo AML Ph1+, 6 myeloid, and three biphenotypic leukemia showed that BCR-ABL1+AML blasts often are burdened by aberration commonly associated with lymphoid lineage tumors (deletions of IKZF and CDKN2A/B and concomitant loss within the immunoglobulin and T cell receptor gene complexes) that are all also found in BCR-ABL1+ ALL and CML chronic phase, but not in myeloid CML-BP.[47]
The presence of lymphoid markers, not sufficient for a classification as MPAL according to WHO, is in line with a finding by Atfy et al., who found in all nine cases of de novo Ph1+ AML: CD33 and CD13 markers, and CD64 in 8 of them. MPO was positive in 9/9 patients by flow cytometry. The B-lymphoid marker CD79a was positive in one, T-lymphoid marker CD7 in 4, CD24 in one case, and CD19 was found in two AML cases that could be considered as FAB M2. Seven of the nine AML patients had an aberrant expression of lymphoid markers. Stem cell markers CD34 were positive in 6/9, and TDT was positive in 1/9 cases. According to the FAB, one case was diagnosed as M0, 3 cases as M1, 4 cases as M2, and one case as M4.[48]
In a recent study among 46 cases of myeloid BP, 76% expressed CD34, and 74% expressed CD117. Myeloperoxidase expression was noted in a variable proportion of precursor cells in 85% of cases. TDT was expressed in 37% cases, 14 cases expressed markers outside of the standard myeloid phenotype, and two expressed markers of more than one lineage (B or/and T).[49]
In a retrospective study of 477 BP cases in 20 years encompassing the introduction of TKI therapy, Jain P et al. found that, for 77 patients diagnosed as BP at the onset, first-line treatment included TKI alone (24 patients; 34%), TKI plus chemotherapy (41 patients; 58%), non-TKI-based therapies (2 patients; 3%). Clonal evolution under therapy pressure must play a role since patients with de novo BP had a longer overall survival time (OS) compared with patients who transformed from CML-Chronic Phase/CML-Acute Phase (P<.0001). The most effective treatment option was the combination of a TKI with chemotherapy. Patients who achieved morphologic hematologic remission (MHR) or complete cytogenetic remission (CCyR) or major molecular response (MMR) after initial BP treatment had a significantly longer failure-free survival (FFS) (P<.0001) and the achievement of MHR and/or CCyR emerged as the most significant independent predictors of survival.[41] In a 2019 review, Soverini et al. state that 2 to 5% of CML patients present in accelerated phase (AP) and 2 to 7% in BP and that, as a whole, AP/BP patients display a high degree of genetic instability, with an accumulation of additional genetic and cytogenetic abnormalities that reduce sensitivity to TKI. However, the paper does not address the genetics of de novo AP/BP patients.[50]
In a study published in 2015, Klco et al. confirm that among 71 patients with de novo AML, 18 patients carrying NPM1 mutated alleles were cleared below the threshold of 2,5% (5% of cells) at day 30 from induction therapy start, and those patients have the best chances to have a long first remission. Seemingly type I mutations as FLT3, KRAS, or NRAS were usually cleared on day 30, suggesting that subclones containing these mutations may be highly sensitive to induction chemotherapy, but of course, those patients tend to relapse early and have a poor prognosis.[51]
Thus would not be unusual that in our case, if considered as an AML, the molecular profile was nearly negative for both mutations on day 45. Conversely, a CML-BP in hematological remission after just one intensive chemotherapy cycle and after no more than 18 days of TKI therapy should register at least a substantial regrowth of clonal BCR-ABL1 positive hematopoiesis. As exposed before, there are several suggestions but not certainty about discriminating de novo Ph1+AML and CML-BP at the onset. In the absence of a previous CML history, differential diagnosis is based on the global analysis of histologic, immunophenotype, and genetic features, which in most cases singularly are not decisive in differentiating the two conditions, but taken all together may lead to a possible assignation. We summarize the differential characteristics in Table 2. It is interesting as some statistical modeling reports implicate the role of functional NPM1 in conveying tumorigenic signals from the BCR-ABL1 oncoprotein to ribosome biogenesis, affecting cellular growth.[52,53] Thus, in theory, NPM1 mutation could hamper, in part, BCR-ABL1 oncogenic phenotype, which explains the rarity of the finding and renders NPM1 a highly improbable candidate for BP transition.


Table 2 Table 2.  Differential characteristics between CML-BP and AML.



Conclusions

In conclusion, there seems to be only one clear precedent of CML-BP carrying the NPM1 mutation, convincing under the clinical point of view since we are given no information about the mutational status of the CP,[64] whereas double mutated NPM1 BCR-ABL1+ AML, although rare, has been clearly devised as part of the AML with mutated NPM1 classification. We feel that NPM1 mutation presence has to be considered decidedly as a sign of AML rather than BP. Therapeutic decisions must consider the broader clinical aspects, the comparatively mild side-effects of TKI therapy versus the great benefit that might bring to most of the patients, as our case history may incidentally demonstrate it. Even considering all the premises, and even after the chemical pleuritis he suffered after five years of Dasatinib at 100 mg per day, a complication promptly resolved with 30 days interruption, diuretic, and corticoid therapy. However, we are not counseling to end TKI therapy; since there are no antecedents to guide us, we rather play safe continuing a course of action that was highly effective and with affordable side effects so far.

References   

  1. Papaemmanuil E, Gerstung M, Bullinger L, Gaidzik VI, Paschka P, Roberts ND et al. Genomic classification and prognosis in acute myeloid leukemia. N Engl J Med. 2016; 374(23):2209-2221. https://doi.org/10.1056/NEJMoa1516192 PMid:27276561 PMCid:PMC4979995
  2. Keung YK, Beaty M, Powell BL, Molnar I, Buss D, Pettenati M. Philadelphia chromosome positive myelodysplastic syndrome and acute myeloid leukemia - Retrospective study and review of literature. Leuk Research 2004; 28(6):579-86. https://doi.org/10.1016/j.leukres.2003.10.027 PMid:15120934
  3. Soupir CP, Vergilio JA, Dal Cin P, Muzikansky A, Kantarjian H, Jones D et al. Philadelphia chromosome-positive acute myeloid leukemia: A rare aggressive leukemia with clinicopathologic features distinct from chronic myeloid leukemia in myeloid blast crisis. Am J Clin Pathol 2007; 127(4): 642-50. https://doi.org/10.1309/B4NVER1AJJ84CTUU PMid:17369142
  4. Cuneo A, Ferrant A, Michaux JL, Demuynck H, Boogaerts M, Louwagie A et al. Philadelphia chromosome-positive acute myeloid leukemia: Cytoimmunologic and cytogenetic features. Haematologica 1996; 81(5):423-7.
  5. Döhner H, Estey E, Grimwade D, Amadori S, Appelbaum FR, Büchner T et al. Diagnosis and management of AML in adults: 2017 ELN recommendations from an international expert panel. Blood 2017; 129(4): 424-447. https://doi.org/10.1182/blood-2016-08-733196 PMid:27895058 PMCid:PMC5291965
  6. Arber DA, Orazi A, Hasserjian R, Thiele J, Borowitz MJ, Le Beau MM et al. The 2016 revision to the World Health Organization classification of myeloid neoplasms and acute leukemia. Blood 2016; 127: 2391-405. https://doi.org/10.1182/blood-2016-03-643544 PMid:27069254
  7. Ley TJ, Miller C, Ding L, Raphael BJ, Mungall AJ, Robertson G et al. Genomic and epigenomic landscapes of adult de novo acute myeloid leukemia. N Engl J Med. 2013; 368: 2059-74. https://doi.org/10.1056/NEJMoa1301689 PMid:23634996 PMCid:PMC3767041
  8. Papayannidis C, Sartor C, Marconi G, Fontana MC, Nanni J, Cristiano G et al. Acute myeloid leukemia mutations: Therapeutic implications. Int J Mol Sci 2019. 20: (11), https://doi.org/10.3390/ijms20112721 PMid:31163594 PMCid:PMC6600275
  9. Noguera NI, Piredda ML, Taulli R, Catalano G, Angelini G, Gaur G et al. PML/RARa inhibits PTEN expression in hematopoietic cells by competing with PU.1 transcriptional activity. Oncotarget 2016, 7: 66386-66397. https://doi.org/10.18632/oncotarget.11964 PMid:27626703 PMCid:PMC5341808
  10. Panuzzo C, Crivellaro S, Carrà G, Guerrasio A, Saglio G, Morotti A. Bcr-abl promotes pten downregulation in chronic myeloid leukemia. PLoS ONE 2014; 9: e110682. https://doi.org/10.1371/journal.pone.0110682 PMid:25343485 PMCid:PMC4208795
  11. Huret J-L, Ahmad M, Arsaban M, Jacquemot-Perbal M-C, Le Berre V, Malo A et al. Atlas of Genetics and Cytogenetics in Oncology and Haematology Staff http://AtlasGeneticsOncology.org Atlas of Genetics and Cytogenetics in Oncology and Haematology. Atlas Genet Cytogenet Oncol Haematol 2015.
  12. Okuwaki M. The structure and functions of NPM1/Nucleophsmin/B23, a multifunctional nucleolar acidic protein. J Biochem. 2008; 143: 441-8. https://doi.org/10.1093/jb/mvm222 PMid:18024471
  13. Yip SP, Siu PM, Leung PHM, Zhao Y, Yung BYM. The multifunctional nucleolar protein nucleophosmin/NPM/B23 and the nucleoplasmin family of proteins. Protein Reviews 2011; 15: 213-252. https://doi.org/10.1007/978-1-4614-0514-6_10 PMCid:PMC7121557
  14. Brown P, McIntyre E, Rau R, Meshinchi S, Lacayo N, Dahl G et al. The incidence and clinical significance of nucleophosmin mutations in childhood AML. Blood 2007; 110:979-85. https://doi.org/10.1182/blood-2007-02-076604 PMid:17440048 PMCid:PMC1924773
  15. Calvo KL, Ojeda MJ, Ammatuna E, Lavorgna S, Ottone T, Targovnik HM et al. Detection of the nucleophosmin gene mutations in acute myelogenous leukemia through RT-PCR and polyacrylamide gel electrophoresis. Eu J Haematol. 2009, 82: 69-72. https://doi.org/10.1111/j.1600-0609.2008.01155.x PMid:18801061
  16. Falini B, Martelli MP, Bolli N, Sportoletti P, Liso A, Tiacci E et al. Acute myeloid leukemia with mutated nucleophosmin (NPM1): Is it a distinct entity? Blood 2011; 117: 1109-20. https://doi.org/10.1182/blood-2010-08-299990 PMid:21030560
  17. Martínez-Losada C, Serrano-López J, Serrano-López J, Noguera NI, Garza E, Piredda L et al. Clonal genetic evolution at relapse of favorable-risk acute myeloid leukemia with NPM1 mutation is associated with phenotypic changes and worse outcomes. Haematologica 2018; 103: e400-e403. https://doi.org/10.3324/haematol.2018.188433 PMid:29622659 PMCid:PMC6119134
  18. Federici L, Falini B. Nucleophosmin mutations in acute myeloid leukemia: A tale of protein unfolding and mislocalization. Protein Sci. 2013; 22: 545-56. https://doi.org/10.1002/pro.2240 PMid:23436734 PMCid:PMC3649256
  19. Noguera NI, Song MS, Divona M, Catalano G, Calvo KL, García F et al. Nucleophosmin/B26 regulates PTEN through interaction with HAUSP in acute myeloid leukemia. Leukemia 2013; 27: 1037-43. https://doi.org/10.1038/leu.2012.314 PMid:23183427
  20. Colombo E, Alcalay M, Pelicci PG. Nucleophosmin and its complex network: A possible therapeutic target in hematological diseases. Oncogene 2011; 30:2595-609. https://doi.org/10.1038/onc.2010.646 PMid:21278791
  21. Colombo E, Bonetti P, Lazzerini Denchi E, Martinelli P, Zamponi R, Marine J-C et al. Nucleophosmin Is Required for DNA Integrity and p19Arf Protein Stability. Mol Cell Biol. 2005; 25(20):8874-86. https://doi.org/10.1128/MCB.25.20.8874-8886.2005 PMid:16199867 PMCid:PMC1265791
  22. Stephens PJ, Greenman CD, Fu B, Yang F, Bignell GR, Mudie LJ et al. Massive genomic rearrangement acquired in a single catastrophic event during cancer development. Cell 2011; 144(1):27-40. https://doi.org/10.1016/j.cell.2010.11.055 PMid:21215367 PMCid:PMC3065307
  23. Fontana MC, Marconi G, Feenstra JDM, Fonzi E, Papayannidis C, Ghelli Luserna Di Rorá A et al. Chromothripsis in acute myeloid leukemia: Biological features and impact on survival. Leukemia 2018; 32:1609-1620. https://doi.org/10.1038/s41375-018-0035-y PMid:29472722 PMCid:PMC6035145
  24. Welch JS, Ley TJ, Link DC, Miller CA, Larson DE, Koboldt DC et al. The origin and evolution of mutations in acute myeloid leukemia. Cell 2012; 150(2):264-78. https://doi.org/10.1016/j.cell.2012.06.023 PMid:22817890 PMCid:PMC3407563
  25. Noguera NI, Catalano G, Banella C, Divona M, Faraoni I, Ottone T et al. Acute promyelocytic Leukemia: Update on the mechanisms of leukemogenesis, resistance and on innovative treatment strategies. Cancers 2019; 11(10):1591. https://doi.org/10.3390/cancers11101591 PMid:31635329 PMCid:PMC6826966
  26. Krönke J, Bullinger L, Teleanu V, Tschürtz F, Gaidzik VI, Kühn MWM et al. Clonal evolution in relapsed NPM1-mutated acute myeloid leukemia. Blood 2013; 122(1):100-8. https://doi.org/10.1182/blood-2013-01-479188 PMid:23704090
  27. Jan M, Snyder TM, Corces-Zimmerman MR, Vyas P, Weissman IL, Quake SR et al. Clonal evolution of preleukemic hematopoietic stem cells precedes human acute myeloid leukemia. Sci Trans Med 2012; 4(149):149ra118. doi:10.1126/scitranslmed.3004315. https://doi.org/10.1126/scitranslmed.3004315 PMid:22932223 PMCid:PMC4045621
  28. Potter N, Miraki-Moud F, Ermini L, Titley I, Vijayaraghavan G, Papaemmanuil E et al. Single cell analysis of clonal architecture in acute myeloid leukaemia. Leukemia 2019; 33(5):1113-1123. https://doi.org/10.1038/s41375-018-0319-2 PMid:30568172 PMCid:PMC6451634
  29. Gilliland DG. Molecular genetics of human leukemias: New insights into therapy. Sem Hematol 2002.; 39:6-11. doi:10.1053/shem.2002.36921. https://doi.org/10.1053/shem.2002.36921 PMid:12447846
  30. Gilliland, D.G.; Griffin, J.D. The roles of FLT3 in hematopoiesis and leukemia. Blood 2002; 39:6-11. https://doi.org/10.1182/blood-2002-02-0492 PMid:12176867
  31. Bacher U, Haferlach T, Alpermann T, Zenger M, Hochhaus A, Beelen DW et al. Subclones with the t(9;22)/BCR-ABL1 rearrangement occur in AML and seem to cooperate with distinct genetic alterations. Br J Haematol; 2011; 152(6):713-20. https://doi.org/10.1111/j.1365-2141.2010.08472.x PMid:21275954
  32. Biernaux C, Loos M, Sels A, Huez G, Stryckmans P. Detection of major bcr-abl gene expression at a very low level in blood cells of some healthy individuals. Blood 1995; 86: 3118-22. https://doi.org/10.1182/blood.V86.8.3118.3118
  33. Ismail SI, Naffa RG, Yousef AMF, Ghanim MT. Incidence of bcr-abl fusion transcripts in healthy individuals. Med Rep. 2014; 9: 1271-6. https://doi.org/10.3892/mmr.2014.1951 PMid:24535287
  34. Suzuki T, Kiyoi H, Ozeki K, Tomita A, Yamaji S, Suzuki R et al. Clinical characteristics and prognostic implications of NPM1 mutations in acute myeloid leukemia. Blood 2005; 106(8):2854-61. https://doi.org/10.1182/blood-2005-04-1733 PMid:15994285
  35. Verhaak RGW, Goudswaard CS, Van Putten W, Bijl MA, Sanders MA, Hugens W et al. Mutations in nucleophosmin (NPM1) in acute myeloid leukemia (AML): Association with other gene abnormalities and previously established gene expression signatures and their favorable prognostic significance. Blood 2005; 106: 3747-54. https://doi.org/10.1182/blood-2005-05-2168 PMid:16109776
  36. Konoplev S, Yin CC, Kornblau SM, Kantarjian HM, Konopleva M, Andreeff M et al. Molecular characterization of de novo Philadelphia chromosome-positive acute myeloid leukemia. Leuk Lymphoma 2013; 54: 138-44. https://doi.org/10.3109/10428194.2012.701739 PMid:22691121 PMCid:PMC3925981
  37. Reboursiere E, Chantepie S, Gac AC, Reman O. Rare but authentic Philadelphia-positive acute myeloblastic leukemia: Two case reports and a literature review of characteristics, treatment and outcome. Hematology/ Oncology and Stem Cell Ther. 2014, 8(1):28-33. https://doi.org/10.1016/j.hemonc.2014.09.002 PMid:25300567
  38. Mariotti B, Meconi F, Palmieri R, De Bellis E, Lavorgna S, Ottone T et al. Acute Myeloid Leukemia with Concomitant BCR-ABL and NPM1 Mutations. Case Rep Hematol. 2019; 6707506. https://doi.org/10.1155/2019/6707506 PMid:31110828 PMCid:PMC6487162
  39. Castagnetti F, Gugliotta G, Breccia M, Iurlo A, Levato L, Albano F et al. The BCR-ABL1 transcript type influences response and outcome in Philadelphia chromosome-positive chronic myeloid leukemia patients treated frontline with imatinib. Am J Hematol. 2017; 92(8):797-805. https://doi.org/10.1002/ajh.24774 PMid:28466557
  40. Neuendorff NR, Burmeister T, Dörken B, Westermann J. BCR-ABL-positive acute myeloid leukemia: a new entity? Analysis of clinical and molecular features. Ann Hematol. 2016; 95: 1211-21. https://doi.org/10.1007/s00277-016-2721-z PMid:27297971
  41. Jain P, Kantarjian HM, Ghorab A, Sasaki K, Jabbour EJ, Nogueras Gonzalez G et al. Prognostic factors and survival outcomes in patients with chronic myeloid leukemia in blast phase in the tyrosine kinase inhibitor era: Cohort study of 477 patients. Cancer 2017; 123(22):4391-4402. https://doi.org/10.1002/cncr.30864 PMid:28743165 PMCid:PMC5673547
  42. Gabert J, Beillard E, van der Velden VHJ, Bi W, Grimwade D, Pallisgaard N et 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: 2318-2357. https://doi.org/10.1038/sj.leu.2403135 PMid:14562125
  43. Ammatuna E, Noguera NI, Zangrilli D, Curzi P, Panetta P, Bencivenga P et al. Rapid detection of nucleophosmin (NPM1) mutations in acute myeloid leukemia by denaturing HPLC. Clin Chem 2005, 51: 2165-2167. https://doi.org/10.1373/clinchem.2005.055707 PMid:16244291
  44. Gorello P, Cazzaniga G, Alberti F, Dell' Oro MG, Gottardi E, Specchia G et al. Quantitative assessment of minimal residual disease in acute myeloid leukemia carrying nucleophosmin (NPM1) gene mutations. Leukemia 2006; 20: 1103-1108. https://doi.org/10.1038/sj.leu.2404149 PMid:16541144
  45. Reid AG, De Melo VA, Elderfield K, Clark I, Marin D, Apperley J et al. phenotype of blasts in chronic myeloid leukemia in blastic phase-Analysis of bone marrow trephine biopsies and correlation with cytogenetics. Leuk Research 2009, 33: 418-425. https://doi.org/10.1016/j.leukres.2008.07.019 PMid:18760473
  46. Shah NP, Nicoll JM, Nagar B, Gorre ME, Paquette RL, Kuriyan J et al. Multiple BCR-ABL kinase domain mutations confer polyclonal resistance to the tyrosine kinase inhibitor imatinib (STI571) in chronic phase and blast crisis chronic myeloid leukemia. Cancer Cell 2002; 2: 117-25. https://doi.org/10.1016/S1535-6108(02)00096-X
  47. Nacheva EP, Grace CD, Brazma D, Gancheva K, Howard-Reeves J, Rai L et al. Does BCR/ABL1 positive Acute Myeloid Leukaemia Exist?. Br J Haematol. 2013; 161: 541-50. https://doi.org/10.1111/bjh.12301 PMid:23521501
  48. Atfy M, Al Azizi NMA, Elnaggar AM. Incidence of Philadelphia-chromosome in acute myelogenous leukemia and biphenotypic acute leukemia patients: And its role in their outcome. Leuk Research 2011. 35: 1339-44. https://doi.org/10.1016/j.leukres.2011.04.011 PMid:21612824
  49. Varma S, Varma N, Malhotra P, Narang V, Sachdeva MU, Bose P. Immunophenotyping in Chronic Myeloid Leukemia Blast Crisis: Looking beyond Morphology. J Postgrad Med Edu Res. 2016; 50: 181-184.52. https://doi.org/10.5005/jp-journals-10028-1215
  50. Soverini S, Bassan R, Lion T. Treatment and monitoring of Philadelphia chromosome-positive leukemia patients: Recent advances and remaining challenges. J Hematol Oncol. 2019;12(1):39. https://doi.org/10.1186/s13045-019-0729-2 PMid:31014376 PMCid:PMC6480772
  51. Klco JM, Miller CA, Griffith M, Petti A, Spencer DH, Ketkar-Kulkarni S et al. Association between mutation clearance after induction therapy and outcomes in acute myeloid leukemia. JAMA 2015; 314: 811-822. https://doi.org/10.1001/jama.2015.9643 PMid:26305651 PMCid:PMC4621257
  52. Wang F, Chan LWC, Tsui NBY, Wong SCC, Siu PM, Yip SP et al. Coexpression pattern analysis of NPM1-associated genes in chronic myelogenous leukemia. Biomed Res Int. 2015; 2015: 1-9. https://doi.org/10.1155/2015/610595 PMid:25961029 PMCid:PMC4413041
  53. Chan LWC, Lin X, Yung G, Lui T, Chiu YM, Wang F et al. Novel structural co-expression analysis linking the NPM1-associated ribosomal biogenesis network to chronic myelogenous leukemia. Sci Rep 2015; 5: 10973. https://doi.org/10.1038/srep10973 PMid:26205693 PMCid:PMC4513283
  54. Quintás-Cardama A, Kantarjian H, Cortes J. Basophilic Blast Phase (B-BP) of Chronic Myelogenous Leukemia (CML). Blood 2007; 110: 4562. https://doi.org/10.1182/blood.V110.11.4562.4562  
  55. Sawyers CL. Chronic myeloid leukemia. N Engl J Med. 1999; 340:1330-40. https://doi.org/10.1056/NEJM199904293401706 PMid:10219069
  56. Kadam PR, Nanjangud GJ, Advani SH, Nair C, Banavali S, Gopal R et al. Chromosomal characteristics of chronic and blastic phase of chronic myeloid leukemia. A study of 100 patients in India. Cancer Genet Cytogenet. 1991; 51:167-81. https://doi.org/10.1016/0165-4608(91)90129-I
  57. Khalidi HS, Brynes RK, Medeiros LJ, Chang KL, Slovak ML, Snyder DS et al. The immunophenotype of blast transformation of chronic myelogenous leukemia: A high frequency of mixed lineage phenotype in 'lymphoid' blasts and a comparison of morphologic, immunophenotypic, and molecular findings. Mod Pathol. 1998; 11: 1211-21.
  58. Branford S, Kim DDH, Apperley JF, Eide CA, Mustjoki S, Ong ST et al. Laying the foundation for genomically-based risk assessment in chronic myeloid leukemia. Leukemia. 2019; 33: 1835-1850. https://doi.org/10.1038/s41375-019-0512-y PMid:31209280 PMCid:PMC6893870
  59. Moarii M, Papaemmanuil E. Classification and risk assessment in AML: Integrating cytogenetics and molecular profiling. 2017; 2017: 37-44. https://doi.org/10.1182/asheducation-2017.1.37 PMid:29222235 PMCid:PMC6142605
  60. Patel JP, Gönen M, Figueroa ME, Fernandez H, Sun Z, Racevskis J et al. Prognostic relevance of integrated genetic profiling in acute myeloid leukemia. N Engl J Med. 2012; 366: 1079-89. https://doi.org/10.1056/NEJMoa1112304 PMid:22417203 PMCid:PMC3545649
  61. Vitale C, Lu X, Abderrahman B, Takahashi K, Ravandi F, Jabbour E. t(9;22) as secondary alteration in core-binding factor de novo acute myeloid leukemia. Am J Hematol 2015; 90:E211-2. https://doi.org/10.1002/ajh.24143 PMid:26257212 PMCid:PMC4807602
  62. Salem A, Loghavi S, Tang G, Huh YO, Jabbour EJ, Kantarjian H et al. Myeloid neoplasms with concurrent BCR-ABL1 and CBFB rearrangements: A series of 10 cases of a clinically aggressive neoplasm. Am J Hematol. 2017; 92: 520-528. https://doi.org/10.1002/ajh.24710 PMid:28253536 PMCid:PMC5860811
  63. Lessard M, Le Prisé PY. Cytogenetic studies in 56 cases with Ph1-positive hematologic disorders. Cancer Genet Cytogenet. 1982; 5: 37-49. https://doi.org/10.1016/0165-4608(82)90039-5
  64. Piccaluga PP, Sabattini E, Bacci F, Agostinelli C, Righi S, Salmi F et al. Cytoplasmic mutated nucleophosmin (NPM1) in blast crisis of chronic myeloid leukaemia. Leukemia 2009; 23: 1370-1. https://doi.org/10.1038/leu.2009.95 PMid:19421226

[TOP]