114 results on '"Falini, B."'
Search Results
2. Criteria for Diagnosis and Molecular Monitoring of NPM1-Mutated AML.
- Author
-
Falini B and Dillon R
- Subjects
- Adult, Humans, Nucleophosmin, Mutation, Risk Factors, Nuclear Proteins genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
NPM1-mutated acute myeloid leukemia (AML) represents the largest molecular subgroup of adult AML. NPM1-mutated AML is recognizable by molecular techniques and immunohistochemistry, which, when combined, can solve difficult diagnostic problems (including identification of myeloid sarcoma and NPM1 mutations outside exon 12). According to updated 2022 European LeukemiaNet (ELN) guidelines, determining the mutational status of NPM1 (and FLT3) is a mandatory step for the genetic-based risk stratification of AML. Monitoring of measurable residual disease (MRD) by qRT-PCR, combined with ELN risk stratification, can guide therapeutic decisions at the post-remission stage. Here, we review the criteria for appropriate diagnosis and molecular monitoring of NPM1-mutated AML., Significance: NPM1-mutated AML represents a distinct entity in the 2022 International Consensus Classification and 5th edition of World Health Organization classifications of myeloid neoplasms. The correct diagnosis of NPM1-mutated AML and its distinction from other AML entities is extremely important because it has clinical implications for the management of AML patients, such as genetic-based risk stratification according to 2022 ELN. Monitoring of MRD by qRT-PCR, combined with ELN risk stratification, can guide therapeutic decisions at the post-remission stage, e.g., whether or not to perform allogeneic hematopoietic stem cell transplantation., (©2023 American Association for Cancer Research.)
- Published
- 2024
- Full Text
- View/download PDF
3. The NPM1 mutant defines AML irrespective of blast count.
- Author
-
Falini B, Martelli MP, Brunetti L, Gjertsen BT, and Andresen V
- Subjects
- Humans, Nucleophosmin, Mutation, fms-Like Tyrosine Kinase 3, Nuclear Proteins genetics, Leukemia, Myeloid, Acute
- Published
- 2023
- Full Text
- View/download PDF
4. NPM1-mutated AML with starry sky pattern.
- Author
-
Falini B, Cardinali V, Brunetti L, and Martelli MP
- Subjects
- Humans, Mutation, Nucleophosmin, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics
- Published
- 2023
- Full Text
- View/download PDF
5. Overlapping features of therapy-related and de novo NPM1-mutated AML.
- Author
-
Othman J, Meggendorfer M, Tiacci E, Thiede C, Schlenk R, Dillon R, Stasik S, Venanzi A, Bertoli S, Delabesse E, Dumas PY, Pigneux A, Bidet A, Gilkes AF, Thomas I, Voso MT, Rambaldi A, Brunetti L, Perriello VM, Andresen V, Gjertsen BT, Martelli MP, Récher C, Röllig C, Bornhäuser M, Serve H, Müller-Tidow C, Baldus CD, Haferlach T, Russell N, and Falini B
- Subjects
- Humans, Nucleophosmin, Mutation, Prognosis, Nuclear Proteins genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute therapy
- Abstract
NPM 1-mutated acute myeloid leukemia (AML) shows unique features. However, the characteristics of "therapy-related" NPM1-mutated AML (t-NPM1 AML) are poorly understood. We compared the genetics, transcriptional profile, and clinical outcomes of t-NPM1 AML, de novo NPM1-mutated AML (dn-NPM1 AML), and therapy-related AML (t-AML) with wild-type NPM1 (t-AML). Normal karyotype was more frequent in t-NPM1 AML (n = 78/96, 88%) and dn-NPM1 (n = 1986/2394, 88%) than in t-AML (n = 103/390, 28%; P < .001). DNMT3A and TET2 were mutated in 43% and 40% of t-NPM1 AML (n = 107), similar to dn-NPM1 (n = 88, 48% and 30%; P > 0.1), but more frequently than t-AML (n = 162; 14% and 10%; P < 0.001). Often mutated in t-AML, TP53 and PPM1D were wild-type in 97% and 96% of t-NPM1 AML, respectively. t-NPM1 and dn-NPM1 AML were transcriptionally similar, (including HOX genes upregulation). At 62 months of median follow-up, the 3-year overall survival (OS) for t-NPM1 AML (n = 96), dn-NPM1 AML (n = 2394), and t-AML (n = 390) were 54%, 60%, and 31%, respectively. In multivariable analysis, OS was similar for the NPM1-mutated groups (hazard ratio [HR] 0.9; 95% confidence interval [CI], 0.65-1.25; P = .45), but better in t-NPM1 AML than in t-AML (HR, 1.86; 95% CI, 1.30-2.68; P < .001). Relapse-free survival was similar between t-NPM1 and dn-NPM1 AML (HR, 1.02; 95% CI, 0.72-1.467; P = .90), but significantly higher in t-NPM1 AML versus t-AML (HR, 1.77; 95% CI, 1.19-2.64; P = .0045). t-NPM1 and dn-NPM1 AML have overlapping features, suggesting that they should be classified as a single disease entity., (© 2023 by The American Society of Hematology.)
- Published
- 2023
- Full Text
- View/download PDF
6. Current status and future perspectives in targeted therapy of NPM1-mutated AML.
- Author
-
Ranieri R, Pianigiani G, Sciabolacci S, Perriello VM, Marra A, Cardinali V, Pierangeli S, Milano F, Gionfriddo I, Brunetti L, Martelli MP, and Falini B
- Subjects
- Adult, Dactinomycin therapeutic use, Histones genetics, Humans, Mutation, Nucleophosmin, Proto-Oncogene Proteins c-bcl-2 metabolism, RNA, Receptors, Antigen, T-Cell genetics, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Nuclear Proteins metabolism
- Abstract
Nucleophosmin 1 (NPM1) is a nucleus-cytoplasmic shuttling protein which is predominantly located in the nucleolus and exerts multiple functions, including regulation of centrosome duplication, ribosome biogenesis and export, histone assembly, maintenance of genomic stability and response to nucleolar stress. NPM1 mutations are the most common genetic alteration in acute myeloid leukemia (AML), detected in about 30-35% of adult AML and more than 50% of AML with normal karyotype. Because of its peculiar molecular and clinico-pathological features, including aberrant cytoplasmic dislocation of the NPM1 mutant and wild-type proteins, lack of involvement in driving clonal hematopoiesis, mutual exclusion with recurrent cytogenetic abnormalities, association with unique gene expression and micro-RNA profiles and high stability at relapse, NPM1-mutated AML is regarded as a distinct genetic entity in the World Health Organization (WHO) classification of hematopoietic malignancies. Starting from the structure and functions of NPM1, we provide an overview of the potential targeted therapies against NPM1-mutated AML and discuss strategies aimed at interfering with the oligomerization (compound NSC348884) and the abnormal traffic of NPM1 (avrainvillamide, XPO1 inhibitors) as well as at inducing selective NPM1-mutant protein degradation (ATRA/ATO, deguelin, (-)-epigallocatechin-3-gallate, imidazoquinoxaline derivatives) and at targeting the integrity of nucleolar structure (actinomycin D). We also discuss the current therapeutic results obtained in NPM1-mutated AML with the BCL-2 inhibitor venetoclax and the preliminary clinical results using menin inhibitors targeting HOX/MEIS1 expression. Finally, we review various immunotherapeutic approaches in NPM1-mutated AML, including immune check-point inhibitors, CAR and TCR T-cell-based therapies against neoantigens created by the NPM1 mutations., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
7. Actinomycin D Targets NPM1c-Primed Mitochondria to Restore PML-Driven Senescence in AML Therapy.
- Author
-
Wu HC, Rérolle D, Berthier C, Hleihel R, Sakamoto T, Quentin S, Benhenda S, Morganti C, Wu C, Conte L, Rimsky S, Sebert M, Clappier E, Souquere S, Gachet S, Soulier J, Durand S, Trowbridge JJ, Bénit P, Rustin P, El Hajj H, Raffoux E, Ades L, Itzykson R, Dombret H, Fenaux P, Espeli O, Kroemer G, Brunetti L, Mak TW, Lallemand-Breitenbach V, Bazarbachi A, Falini B, Ito K, Martelli MP, and de Thé H
- Subjects
- Dactinomycin pharmacology, Dactinomycin therapeutic use, Humans, Mitochondria metabolism, Nucleophosmin, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Nuclear Proteins genetics, Nuclear Proteins metabolism
- Abstract
Acute myeloid leukemia (AML) pathogenesis often involves a mutation in the NPM1 nucleolar chaperone, but the bases for its transforming properties and overall association with favorable therapeutic responses remain incompletely understood. Here we demonstrate that an oncogenic mutant form of NPM1 (NPM1c) impairs mitochondrial function. NPM1c also hampers formation of promyelocytic leukemia (PML) nuclear bodies (NB), which are regulators of mitochondrial fitness and key senescence effectors. Actinomycin D (ActD), an antibiotic with unambiguous clinical efficacy in relapsed/refractory NPM1c-AMLs, targets these primed mitochondria, releasing mitochondrial DNA, activating cyclic GMP-AMP synthase signaling, and boosting reactive oxygen species (ROS) production. The latter restore PML NB formation to drive TP53 activation and senescence of NPM1c-AML cells. In several models, dual targeting of mitochondria by venetoclax and ActD synergized to clear AML and prolong survival through targeting of PML. Our studies reveal an unexpected role for mitochondria downstream of NPM1c and implicate a mitochondrial/ROS/PML/TP53 senescence pathway as an effector of ActD-based therapies., Significance: ActD induces complete remissions in NPM1-mutant AMLs. We found that NPM1c affects mitochondrial biogenesis and PML NBs. ActD targets mitochondria, yielding ROS which enforce PML NB biogenesis and restore senescence. Dual targeting of mitochondria with ActD and venetoclax sharply potentiates their anti-AML activities in vivo. This article is highlighted in the In This Issue feature, p. 2945., (©2021 The Authors; Published by the American Association for Cancer Research.)
- Published
- 2021
- Full Text
- View/download PDF
8. Dactinomycin induces complete remission associated with nucleolar stress response in relapsed/refractory NPM1-mutated AML.
- Author
-
Gionfriddo I, Brunetti L, Mezzasoma F, Milano F, Cardinali V, Ranieri R, Venanzi A, Pierangeli S, Vetro C, Spinozzi G, Dorillo E, Wu HC, Berthier C, Ciurnelli R, Griffin MJ, Jennings CE, Tiacci E, Sportoletti P, Falzetti F, de Thé H, Veal GJ, Martelli MP, and Falini B
- Subjects
- Aged, Antibiotics, Antineoplastic therapeutic use, Cell Nucleolus pathology, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Male, Middle Aged, Neoplasm Recurrence, Local genetics, Neoplasm Recurrence, Local pathology, Nucleophosmin, Pilot Projects, Prognosis, Remission Induction, Salvage Therapy, Cell Nucleolus drug effects, Dactinomycin therapeutic use, Drug Resistance, Neoplasm, Leukemia, Myeloid, Acute drug therapy, Mutation, Neoplasm Recurrence, Local drug therapy, Nuclear Proteins genetics
- Abstract
Acute myeloid leukemia (AML) with mutated NPM1 accounts for one-third of newly diagnosed AML. Despite recent advances, treatment of relapsed/refractory NPM1-mutated AML remains challenging, with the majority of patients eventually dying due to disease progression. Moreover, the prognosis is particularly poor in elderly and unfit patients, mainly because they cannot receive intensive treatment. Therefore, alternative treatment strategies are needed. Dactinomycin is a low-cost chemotherapeutic agent, which has been anecdotally reported to induce remission in NPM1-mutated patients, although its mechanism of action remains unclear. Here, we describe the results of a single-center phase 2 pilot study investigating the safety and efficacy of single-agent dactinomycin in relapsed/refractory NPM1-mutated adult AML patients, demonstrating that this drug can induce complete responses and is relatively well tolerated. We also provide evidence that the activity of dactinomycin associates with nucleolar stress both in vitro and in vivo in patients. Finally, we show that low-dose dactinomycin generates more efficient stress response in cells expressing NPM1 mutant compared to wild-type cells, suggesting that NPM1-mutated AML may be more sensitive to nucleolar stress. In conclusion, we establish that dactinomycin is a potential therapeutic alternative in relapsed/refractory NPM1-mutated AML that deserves further investigation in larger clinical studies., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
9. How I diagnose and treat NPM1-mutated AML.
- Author
-
Falini B, Brunetti L, and Martelli MP
- Subjects
- Age Factors, Aged, Algorithms, Allografts, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow pathology, Bridged Bicyclo Compounds, Heterocyclic administration & dosage, Cell Lineage, Clinical Trials as Topic, Clonal Evolution, Combined Modality Therapy, Diagnosis, Differential, Disease Management, Female, Gemtuzumab administration & dosage, Hematopoietic Stem Cell Transplantation, Humans, Leukemia, Myeloid, Acute classification, Leukemia, Myeloid, Acute genetics, Male, Middle Aged, Molecular Targeted Therapy, Myelodysplastic Syndromes chemically induced, Myelodysplastic Syndromes diagnosis, Neoplastic Stem Cells pathology, Nuclear Proteins antagonists & inhibitors, Nucleophosmin, Oncogene Proteins, Fusion antagonists & inhibitors, Patient Selection, Remission Induction, Risk Assessment, Salvage Therapy, Sulfonamides administration & dosage, fms-Like Tyrosine Kinase 3 genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute therapy, Nuclear Proteins genetics, Oncogene Proteins, Fusion genetics, Practice Patterns, Physicians'
- Abstract
Mutations of the nucleophosmin (NPM1) gene, encoding for a nucleolar multifunctional protein, occur in approximately one-third of adult acute myeloid leukemia (AML). NPM1-mutated AML exhibits unique molecular, pathological, and clinical features, which led to its recognition as distinct entity in the 2017 World Health Organization (WHO) classification of myeloid neoplasms. Although WHO criteria for the diagnosis of NPM1-mutated AML are well established, its distinction from other AML entities may be difficult. Moreover, the percentage of blasts required to diagnose NPM1-mutated AML remains controversial. According to the European LeukemiaNet (ELN), determining the mutational status of NPM1 (together with FLT3) is mandatory for accurate relapse-risk assessment. NPM1 mutations are ideal targets for measurable residual disease (MRD) monitoring, since they are AML specific, frequent, very stable at relapse, and do not drive clonal hematopoiesis of undetermined significance. MRD monitoring by quantitative polymerase chain reaction of NPM1-mutant transcripts, possibly combined with ELN genetic-based risk stratification, can guide therapeutic decisions after remission. Furthermore, immunohistochemistry can be very useful in selected situations, such as diagnosis of NPM1-mutated myeloid sarcoma. Herein, we present 4 illustrative cases of NPM1-mutated AML that address important issues surrounding the biology, diagnosis, and therapy of this common form of leukemia., (© 2021 by The American Society of Hematology.)
- Published
- 2021
- Full Text
- View/download PDF
10. NPM1-mutated acute myeloid leukemia: from bench to bedside.
- Author
-
Falini B, Brunetti L, Sportoletti P, and Martelli MP
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Clonal Hematopoiesis genetics, Disease Management, Humans, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute therapy, Nucleophosmin, Precancerous Conditions genetics, Precancerous Conditions metabolism, Prognosis, Translational Research, Biomedical, Genetic Association Studies, Genetic Predisposition to Disease, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics
- Abstract
The nucleophosmin (NPM1) gene encodes for a multifunctional protein with prominent nucleolar localization that shuttles between nucleus and cytoplasm. NPM1 mutations represent the most common genetic lesion in adult acute myeloid leukemia (AML; about one third of cases), and they act deterministically to cause the aberrant cytoplasmic delocalization of NPM1 mutants. Because of its unique features, NPM1-mutated AML is recognized as a distinct entity in the 2017 World Health Organization (WHO) classification of hematopoietic neoplasms. Here, we focus on recently identified functions of wild-type NPM1 in the nucleolus and address new biological and clinical issues related to NPM1-mutated AML. The relevance of the cooperation between NPM1 and other mutations in driving AML with different outcomes is presented. We also discuss the importance of eradicating NPM1-mutated clones to achieve AML cure and the impact of preleukemic clonal hematopoiesis persistence in predisposing to second AML. The contribution of HOX genes' expression to the development of NPM1-mutated AML is also highlighted. Clinically, yet unsolved diagnostic issues in the 2017 WHO classification of myeloid neoplasms and the importance of NPM1 mutations in defining the framework of European LeukemiaNet genetic-based risk stratification are discussed. Finally, we address the value and limits of NPM1-based measurable residual disease assessment for treatment guidance and present the results of promising preclinical studies with XPO1 and menin-MLL inhibitors., (© 2020 by The American Society of Hematology.)
- Published
- 2020
- Full Text
- View/download PDF
11. Germline NPM1 mutations lead to altered rRNA 2'-O-methylation and cause dyskeratosis congenita.
- Author
-
Nachmani D, Bothmer AH, Grisendi S, Mele A, Bothmer D, Lee JD, Monteleone E, Cheng K, Zhang Y, Bester AC, Guzzetti A, Mitchell CA, Mendez LM, Pozdnyakova O, Sportoletti P, Martelli MP, Vulliamy TJ, Safra M, Schwartz S, Luzzatto L, Bluteau O, Soulier J, Darnell RB, Falini B, Dokal I, Ito K, Clohessy JG, and Pandolfi PP
- Subjects
- Animals, Dyskeratosis Congenita pathology, Gene Expression Profiling, Hematopoietic Stem Cells, Male, Methylation, Mice, Mice, Inbred C57BL, Mice, Knockout, Nuclear Proteins chemistry, Nucleophosmin, RNA, Small Nucleolar, Transcriptome, Dyskeratosis Congenita genetics, Epigenomics methods, Germ-Line Mutation, Nuclear Proteins genetics, RNA Processing, Post-Transcriptional, RNA, Messenger genetics, RNA, Ribosomal genetics
- Abstract
RNA modifications are emerging as key determinants of gene expression. However, compelling genetic demonstrations of their relevance to human disease are lacking. Here, we link ribosomal RNA 2'-O-methylation (2'-O-Me) to the etiology of dyskeratosis congenita. We identify nucleophosmin (NPM1) as an essential regulator of 2'-O-Me on rRNA by directly binding C/D box small nucleolar RNAs, thereby modulating translation. We demonstrate the importance of 2'-O-Me-regulated translation for cellular growth, differentiation and hematopoietic stem cell maintenance, and show that Npm1 inactivation in adult hematopoietic stem cells results in bone marrow failure. We identify NPM1 germline mutations in patients with dyskeratosis congenita presenting with bone marrow failure and demonstrate that they are deficient in small nucleolar RNA binding. Mice harboring a dyskeratosis congenita germline Npm1 mutation recapitulate both hematological and nonhematological features of dyskeratosis congenita. Thus, our findings indicate that impaired 2'-O-Me can be etiological to human disease.
- Published
- 2019
- Full Text
- View/download PDF
12. Getting away with phase transition: NPM1-mutated bone myeloid sarcoma mimicking Ewing sarcoma.
- Author
-
Marra A, Martino G, Ascani S, Martelli MP, and Falini B
- Subjects
- Allografts, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Bone Marrow Transplantation, Bone Neoplasms genetics, Bone Neoplasms pathology, Bone Neoplasms therapy, Combined Modality Therapy, Diagnosis, Differential, Fatal Outcome, Female, Humans, Leukemia, Myeloid, Acute diagnosis, Nucleophosmin, Salvage Therapy, Sarcoma, Myeloid genetics, Sarcoma, Myeloid pathology, Sarcoma, Myeloid therapy, Young Adult, Bone Neoplasms diagnosis, Neoplasm Proteins genetics, Nuclear Proteins genetics, Sarcoma, Ewing diagnosis, Sarcoma, Myeloid diagnosis
- Published
- 2019
- Full Text
- View/download PDF
13. GATA1 epigenetic deregulation contributes to the development of AML with NPM1 and FLT3-ITD cooperating mutations.
- Author
-
Sportoletti P, Celani L, Varasano E, Rossi R, Sorcini D, Rompietti C, Strozzini F, Del Papa B, Guarente V, Spinozzi G, Cecchini D, Bereshchenko O, Haferlach T, Martelli MP, Falzetti F, and Falini B
- Subjects
- Animals, Female, GATA1 Transcription Factor physiology, Gene Expression Regulation, Neoplastic, Humans, Leukemia, Myeloid, Acute etiology, Leukemia, Myeloid, Acute genetics, Mice, Mice, Knockout, Nucleophosmin, Prognosis, Tandem Repeat Sequences, fms-Like Tyrosine Kinase 3 physiology, Biomarkers, Tumor genetics, Epigenesis, Genetic, GATA1 Transcription Factor genetics, Leukemia, Myeloid, Acute pathology, Mutation, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Published
- 2019
- Full Text
- View/download PDF
14. IDH1-R132 changes vary according to NPM1 and other mutations status in AML.
- Author
-
Falini B, Spinelli O, Meggendorfer M, Martelli MP, Bigerna B, Ascani S, Stein H, Rambaldi A, and Haferlach T
- Subjects
- Humans, Nucleophosmin, Prognosis, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mutation, Nuclear Proteins genetics
- Published
- 2019
- Full Text
- View/download PDF
15. Leukemogenic nucleophosmin mutation disrupts the transcription factor hub that regulates granulomonocytic fates.
- Author
-
Gu X, Ebrahem Q, Mahfouz RZ, Hasipek M, Enane F, Radivoyevitch T, Rapin N, Przychodzen B, Hu Z, Balusu R, Cotta CV, Wald D, Argueta C, Landesman Y, Martelli MP, Falini B, Carraway H, Porse BT, Maciejewski J, Jha BK, and Saunthararajah Y
- Subjects
- Animals, Granulocytes pathology, Heterografts, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mice, Monocytes pathology, Neoplasm Proteins genetics, Neoplasm Transplantation, Nuclear Proteins genetics, Nucleophosmin, THP-1 Cells, Transcription Factors genetics, Granulocytes metabolism, Leukemia, Myeloid, Acute metabolism, Monocytes metabolism, Mutation, Neoplasm Proteins metabolism, Nuclear Proteins metabolism, Transcription Factors metabolism
- Abstract
Nucleophosmin (NPM1) is among the most frequently mutated genes in acute myeloid leukemia (AML). It is not known, however, how the resulting oncoprotein mutant NPM1 is leukemogenic. To reveal the cellular machinery in which NPM1 participates in myeloid cells, we analyzed the endogenous NPM1 protein interactome by mass spectrometry and discovered abundant amounts of the master transcription factor driver of monocyte lineage differentiation PU.1 (also known as SPI1). Mutant NPM1, which aberrantly accumulates in cytoplasm, dislocated PU.1 into cytoplasm with it. CEBPA and RUNX1, the master transcription factors that collaborate with PU.1 to activate granulomonocytic lineage fates, remained nuclear; but without PU.1, their coregulator interactions were toggled from coactivators to corepressors, repressing instead of activating more than 500 granulocyte and monocyte terminal differentiation genes. An inhibitor of nuclear export, selinexor, by locking mutant NPM1/PU.1 in the nucleus, activated terminal monocytic fates. Direct depletion of the corepressor DNA methyltransferase 1 (DNMT1) from the CEBPA/RUNX1 protein interactome using the clinical drug decitabine activated terminal granulocytic fates. Together, these noncytotoxic treatments extended survival by more than 160 days versus vehicle in a patient-derived xenotransplant model of NPM1/FLT3-mutated AML. In sum, mutant NPM1 represses monocyte and granulocyte terminal differentiation by disrupting PU.1/CEBPA/RUNX1 collaboration, a transforming action that can be reversed by pharmacodynamically directed dosing of clinical small molecules.
- Published
- 2018
- Full Text
- View/download PDF
16. Mutant NPM1 Maintains the Leukemic State through HOX Expression.
- Author
-
Brunetti L, Gundry MC, Sorcini D, Guzman AG, Huang YH, Ramabadran R, Gionfriddo I, Mezzasoma F, Milano F, Nabet B, Buckley DL, Kornblau SM, Lin CY, Sportoletti P, Martelli MP, Falini B, and Goodell MA
- Subjects
- Aged, Animals, Cell Differentiation genetics, Cell Line, Tumor, Cell Nucleus metabolism, Cytoplasm metabolism, Down-Regulation, Female, Humans, Hydrazines pharmacology, Karyopherins antagonists & inhibitors, Karyopherins metabolism, Leukemia, Myeloid, Acute mortality, Leukemia, Myeloid, Acute pathology, Mice, Mutation, Nuclear Proteins metabolism, Nucleophosmin, Proteolysis, Receptors, Cytoplasmic and Nuclear antagonists & inhibitors, Receptors, Cytoplasmic and Nuclear metabolism, Triazoles pharmacology, Xenograft Model Antitumor Assays, Exportin 1 Protein, Gene Expression Regulation, Leukemic, Homeodomain Proteins metabolism, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics
- Abstract
NPM1 is the most frequently mutated gene in cytogenetically normal acute myeloid leukemia (AML). In AML cells, NPM1 mutations result in abnormal cytoplasmic localization of the mutant protein (NPM1c); however, it is unknown whether NPM1c is required to maintain the leukemic state. Here, we show that loss of NPM1c from the cytoplasm, either through nuclear relocalization or targeted degradation, results in immediate downregulation of homeobox (HOX) genes followed by differentiation. Finally, we show that XPO1 inhibition relocalizes NPM1c to the nucleus, promotes differentiation of AML cells, and prolongs survival of Npm1-mutated leukemic mice. We describe an exquisite dependency of NPM1-mutant AML cells on NPM1c, providing the rationale for the use of nuclear export inhibitors in AML with mutated NPM1., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2018
- Full Text
- View/download PDF
17. High-Risk Clonal Hematopoiesis as the Origin of AITL and NPM1-Mutated AML.
- Author
-
Tiacci E, Venanzi A, Ascani S, Marra A, Cardinali V, Martino G, Codoni V, Schiavoni G, Martelli MP, and Falini B
- Subjects
- Humans, Immunoblastic Lymphadenopathy genetics, Leukemia, Myeloid, Acute pathology, Lymphoma, T-Cell pathology, Male, Middle Aged, Nucleophosmin, Hematopoiesis genetics, Leukemia, Myeloid, Acute genetics, Lymphoma, T-Cell genetics, Mutation, Neoplasms, Second Primary genetics, Nuclear Proteins genetics
- Published
- 2018
- Full Text
- View/download PDF
18. Long non-coding RNA expression profile in cytogenetically normal acute myeloid leukemia identifies a distinct signature and a new biomarker in NPM1-mutated patients.
- Author
-
De Clara E, Gourvest M, Ma H, Vergez F, Tosolini M, Dejean S, Demur C, Delabesse E, Recher C, Touriol C, Martelli MP, Falini B, Brousset P, and Bousquet M
- Subjects
- Apoptosis genetics, Biomarkers, Cell Line, Tumor, Cluster Analysis, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Humans, Karyotype, Leukemia, Myeloid, Acute mortality, Nucleophosmin, Prognosis, Sequence Analysis, RNA, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics, RNA, Long Noncoding genetics, Transcriptome
- Abstract
Long non-coding RNAs are defined as transcripts larger than 200 nucleotides but without protein-coding potential. There is growing evidence of the important role of long non-coding RNAs in cancer initiation, development and progression. In this study, we sought to evaluate the long non-coding RNA expression profile of patients with cytogenetically normal acute myeloid leukemia (AML). RNA-sequencing of 40 cytogenetically normal AML patients allowed us to quantify 11,036 long non-coding RNAs. Among these, more than 8000 were previously undescribed long non-coding RNAs. Using unsupervised analysis, we observed a specific long non-coding RNA expression profile dependent on the mutational status of the NPM1 gene. Statistical analysis allowed us to identify a minimal set of 12 long non-coding RNAs capable of discriminating NPM1 -mutated from NPM1 -wild-type patients. These results were validated by qRT-PCR on an independent cohort composed of 134 cytogenetically normal AML patients. Furthermore, we have identified one putative biomarker, the long non-coding RNA XLOC_109948 whose expression pattern predicts clinical outcome. Interestingly, low XLOC_109948 expression indicates a good prognosis especially for NPM1 -mutated patients. Transient transfection of GapmeR against XLOC_109948 in NPM1 -mutated OCI-AML3 cell line treated with Ara-C or ATRA enhances apoptosis suggesting XLOC_109948 plays a role in drug sensitivity. This study improves our knowledge of the long non-coding RNA transcriptome in cytogenetically normal AML patients. We observed a distinct long non-coding RNA expression profile in patients with the NPM1 mutation. The newly identified XLOC_109948 long non-coding RNA emerged as a strong prognostic factor able to better stratify NPM1-mutated patients., (Copyright© 2017 Ferrata Storti Foundation.)
- Published
- 2017
- Full Text
- View/download PDF
19. Dactinomycin in NPM1-Mutated Acute Myeloid Leukemia.
- Author
-
Falini B, Brunetti L, and Martelli MP
- Subjects
- Bone Marrow Examination, Humans, Leukemia, Myeloid, Acute genetics, Middle Aged, Mutation, Nucleophosmin, Antibiotics, Antineoplastic therapeutic use, Bone Marrow pathology, Dactinomycin therapeutic use, Leukemia, Myeloid, Acute drug therapy, Nuclear Proteins genetics
- Published
- 2015
- Full Text
- View/download PDF
20. Impact of genomics in the clinical management of patients with cytogenetically normal acute myeloid leukemia.
- Author
-
Falini B and Martelli MP
- Subjects
- Antineoplastic Agents therapeutic use, Cytogenetic Analysis, DNA (Cytosine-5-)-Methyltransferases metabolism, DNA Methyltransferase 3A, Gene Expression, Humans, In Situ Hybridization, Fluorescence, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute metabolism, Neoplasm Proteins metabolism, Neoplasm, Residual, Nuclear Proteins metabolism, Nucleophosmin, Patient Selection, Prognosis, Risk, Terminology as Topic, fms-Like Tyrosine Kinase 3 metabolism, DNA (Cytosine-5-)-Methyltransferases genetics, Leukemia, Myeloid, Acute genetics, Mutation, Neoplasm Proteins genetics, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Acute myeloid leukemia (AML) is a clinically and molecularly heterogeneous disease. Cytogenetics and FISH have contributed to the stratification of AML patients into favorable, intermediate, and unfavorable risk categories. However, until recently, the prognostic stratification and treatment decision for the intermediate risk category, mostly comprising AML patients with normal cytogenetics (CN-AML), has been difficult due to the scarce knowledge of the molecular alterations underlying this large AML subgroup (which accounts for about 50% of all adult AML). During the past decade, the discovery of numerous mutations associated with CN-AML has resulted in significant advances in the AML field. Here, we review the biological characteristics of the most common mutations underlying CN-AML and outline their clinical impact in the following settings: (i) definition of new molecular leukemia entities in the WHO classification; (ii) risk stratification of CN-AML patients according to mutational profile; and (iii) monitoring of minimal residual disease by specific quantitative molecular assays., (Copyright © 2015. Published by Elsevier Ltd.)
- Published
- 2015
- Full Text
- View/download PDF
21. Arsenic trioxide and all-trans retinoic acid target NPM1 mutant oncoprotein levels and induce apoptosis in NPM1-mutated AML cells.
- Author
-
Martelli MP, Gionfriddo I, Mezzasoma F, Milano F, Pierangeli S, Mulas F, Pacini R, Tabarrini A, Pettirossi V, Rossi R, Vetro C, Brunetti L, Sportoletti P, Tiacci E, Di Raimondo F, and Falini B
- Subjects
- Animals, Apoptosis genetics, Arsenic Trioxide, Humans, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Mice, Mice, SCID, Mutant Proteins drug effects, Mutant Proteins metabolism, Mutation, Nuclear Proteins drug effects, Nucleophosmin, Oncogene Proteins drug effects, Oncogene Proteins metabolism, Tumor Cells, Cultured, U937 Cells, Xenograft Model Antitumor Assays, Apoptosis drug effects, Arsenicals pharmacology, Leukemia, Myeloid, Acute metabolism, Nuclear Proteins metabolism, Oxides pharmacology, Tretinoin pharmacology
- Abstract
Nucleophosmin (NPM1) mutations represent an attractive therapeutic target in acute myeloid leukemia (AML) because they are common (∼30% AML), stable, and behave as a founder genetic lesion. Oncoprotein targeting can be a successful strategy to treat AML, as proved in acute promyelocytic leukemia by treatment with all-trans retinoic acid (ATRA) plus arsenic trioxide (ATO), which degrade the promyelocytic leukemia (PML)-retinoic acid receptor fusion protein. Adjunct of ATRA to chemotherapy was reported to be beneficial for NPM1-mutated AML patients. Leukemic cells with NPM1 mutation also showed sensibility to ATO in vitro. Here, we explore the mechanisms underlying these observations and show that ATO/ATRA induce proteasome-dependent degradation of NPM1 leukemic protein and apoptosis in NPM1-mutated AML cell lines and primary patients' cells. We also show that PML intracellular distribution is altered in NPM1-mutated AML cells and reverted by arsenic through oxidative stress induction. Interestingly, similarly to what was described for PML, oxidative stress also mediates ATO-induced degradation of the NPM1 mutant oncoprotein. Strikingly, NPM1 mutant downregulation by ATO/ATRA was shown to potentiate response to the anthracyclin daunorubicin. These findings provide experimental evidence for further exploring ATO/ATRA in preclinical NPM1-mutated AML in vivo models and a rationale for exploiting these compounds in chemotherapeutic regimens in clinics., (© 2015 by The American Society of Hematology.)
- Published
- 2015
- Full Text
- View/download PDF
22. Mouse models of NPM1-mutated acute myeloid leukemia: biological and clinical implications.
- Author
-
Sportoletti P, Varasano E, Rossi R, Mupo A, Tiacci E, Vassiliou G, Martelli MP, and Falini B
- Subjects
- Alleles, Animals, Disease Models, Animal, Female, Humans, Leukemia, Myeloid, Acute metabolism, Mice, Mice, Knockout, Mice, SCID, Mice, Transgenic, Mutation, Neoplasm Transplantation, Nuclear Proteins metabolism, Nucleophosmin, Phenotype, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics
- Abstract
Acute myeloid leukemia (AML) carrying nucleophosmin (NPM1) mutations displays distinct biological and clinical features that led to its inclusion as a provisional disease entity in the 2008 World Health Organization (WHO) classification of myeloid neoplasms. Studies of the molecular mechanisms underlying the pathogenesis of NPM1-mutated AML have benefited greatly from several mouse models of this leukemia developed over the past few years. Immunocompromised mice xenografted with NPM1-mutated AML served as the first valuable tool for defining the biology of the disease in vivo. Subsequently, genetically engineered mouse models of the NPM1 mutation, including transgenic and knock-in alleles, allowed the generation of mice with a constant genotype and a reproducible phenotype. These models have been critical for investigating the nature of the molecular effects of these mutations, defining the function of leukemic stem cells in NPM1-mutated AML, identifying chemoresistant preleukemic hemopoietic stem cells and unraveling the key molecular events that cooperate with NPM1 mutations to induce AML in vivo. Moreover, they can serve as a platform for the discovery and validation of new antileukemic drugs in vivo. Advances derived from the analysis of these mouse models promise to greatly accelerate the development of new molecularly targeted therapies for patients with NPM1-mutated AML.
- Published
- 2015
- Full Text
- View/download PDF
23. NPM1 mutations may reveal acute myeloid leukemia in cases otherwise morphologically diagnosed as myelodysplastic syndromes or myelodysplastic/myeloproliferative neoplasms.
- Author
-
Forghieri F, Paolini A, Morselli M, Bigliardi S, Bonacorsi G, Leonardi G, Coluccio V, Maccaferri M, Fantuzzi V, Faglioni L, Colaci E, Soci F, Nasillo V, Messerotti A, Arletti L, Pioli V, Zucchini P, Quadrelli C, Corradini G, Giacobbi F, Vallerini D, Riva G, Barozzi P, Lagreca I, Marasca R, Narni F, Mecucci C, Ottaviani E, Martinelli G, Falini B, Luppi M, and Potenza L
- Subjects
- Aged, Biopsy, Bone Marrow pathology, Cell Transformation, Neoplastic genetics, DNA Mutational Analysis, Female, Humans, Leukemia, Myeloid, Acute diagnosis, Male, Middle Aged, Myelodysplastic Syndromes diagnosis, Myeloproliferative Disorders diagnosis, Nucleophosmin, Leukemia, Myeloid, Acute genetics, Mutation, Myelodysplastic Syndromes genetics, Myeloproliferative Disorders genetics, Nuclear Proteins genetics
- Published
- 2015
- Full Text
- View/download PDF
24. Identification of novel DNA-damage tolerance genes reveals regulation of translesion DNA synthesis by nucleophosmin.
- Author
-
Ziv O, Zeisel A, Mirlas-Neisberg N, Swain U, Nevo R, Ben-Chetrit N, Martelli MP, Rossi R, Schiesser S, Canman CE, Carell T, Geacintov NE, Falini B, Domany E, and Livneh Z
- Subjects
- Cell Line, DNA Repair, DNA-Directed DNA Polymerase genetics, DNA-Directed DNA Polymerase metabolism, Humans, Leukemia, Myeloid, Acute enzymology, Leukemia, Myeloid, Acute genetics, Nuclear Proteins genetics, Nucleophosmin, Protein Binding, Ultraviolet Rays, DNA Damage radiation effects, DNA Replication radiation effects, Leukemia, Myeloid, Acute metabolism, Nuclear Proteins metabolism
- Abstract
Cells cope with replication-blocking lesions via translesion DNA synthesis (TLS). TLS is carried out by low-fidelity DNA polymerases that replicate across lesions, thereby preventing genome instability at the cost of increased point mutations. Here we perform a two-stage siRNA-based functional screen for mammalian TLS genes and identify 17 validated TLS genes. One of the genes, NPM1, is frequently mutated in acute myeloid leukaemia (AML). We show that NPM1 (nucleophosmin) regulates TLS via interaction with the catalytic core of DNA polymerase-η (polη), and that NPM1 deficiency causes a TLS defect due to proteasomal degradation of polη. Moreover, the prevalent NPM1c+ mutation that causes NPM1 mislocalization in ~30% of AML patients results in excessive degradation of polη. These results establish the role of NPM1 as a key TLS regulator, and suggest a mechanism for the better prognosis of AML patients carrying mutations in NPM1.
- Published
- 2014
- Full Text
- View/download PDF
25. A powerful molecular synergy between mutant Nucleophosmin and Flt3-ITD drives acute myeloid leukemia in mice.
- Author
-
Mupo A, Celani L, Dovey O, Cooper JL, Grove C, Rad R, Sportoletti P, Falini B, Bradley A, and Vassiliou GS
- Subjects
- Animals, Cell Transformation, Neoplastic genetics, Disease Models, Animal, Leukemia, Myeloid, Acute metabolism, Mice, Mice, Knockout, Nuclear Proteins metabolism, Nucleophosmin, fms-Like Tyrosine Kinase 3 metabolism, Epistasis, Genetic, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics, Tandem Repeat Sequences, fms-Like Tyrosine Kinase 3 genetics
- Published
- 2013
- Full Text
- View/download PDF
26. Nucleophosmin mutations in acute myeloid leukemia: a tale of protein unfolding and mislocalization.
- Author
-
Federici L and Falini B
- Subjects
- Active Transport, Cell Nucleus, Amino Acid Sequence, Animals, G-Quadruplexes, Gene Expression Regulation, Leukemic, Humans, Models, Molecular, Molecular Sequence Data, Nuclear Proteins metabolism, Nucleophosmin, Protein Folding, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mutation, Nuclear Proteins analysis, Nuclear Proteins genetics
- Abstract
Nucleophosmin (NPM1) is an abundant, ubiquitously expressed protein mainly localized at nucleoli but continuously shuttling between nucleus and cytoplasm. NPM1 plays a role in several cellular functions, including ribosome biogenesis and export, centrosome duplication, chromatin remodeling, DNA repair, and response to stress stimuli. Much of the interest in this protein arises from its relevance in human malignancies. NPM1 is frequently overexpressed in solid tumors and is the target of several chromosomal translocations in hematologic neoplasms. Notably, NPM1 has been characterized as the most frequently mutated gene in acute myeloid leukemia (AML). Mutations alter the C-terminal DNA-binding domain of the protein and result in its aberrant nuclear export and stable cytosolic localization. In this review, we focus on the leukemia-associated NPM1 C-terminal domain and describe its structure, function, and the effect exerted by leukemic mutations. Finally, we discuss the possibility to target NPM1 for the treatment of cancer and, in particular, of AML patients with mutated NPM1 gene., (Copyright © 2013 The Protein Society.)
- Published
- 2013
- Full Text
- View/download PDF
27. The human NPM1 mutation A perturbs megakaryopoiesis in a conditional mouse model.
- Author
-
Sportoletti P, Varasano E, Rossi R, Bereshchenko O, Cecchini D, Gionfriddo I, Bolli N, Tiacci E, Intermesoli T, Zanghì P, Masciulli A, Martelli MP, Falzetti F, Martelli MF, and Falini B
- Subjects
- Animals, Apoptosis, Blotting, Western, Cell Differentiation, Cell Proliferation, Colony-Forming Units Assay, Flow Cytometry, Humans, Immunoenzyme Techniques, Leukemia, Myeloid, Acute pathology, Megakaryocytes metabolism, Mice, Mice, Inbred C57BL, Mice, Transgenic, MicroRNAs genetics, Nucleophosmin, RNA, Messenger genetics, Real-Time Polymerase Chain Reaction, Reverse Transcriptase Polymerase Chain Reaction, Disease Models, Animal, Integrases metabolism, Leukemia, Myeloid, Acute etiology, Megakaryocytes pathology, Mutation genetics, Nuclear Proteins genetics, Thrombopoiesis genetics
- Abstract
The NPM1 mutation is the most frequent genetic alteration thus far identified in acute myeloid leukemia (AML). Despite progress in the clinical and biological characterization of NPM1-mutated AML, the role of NPM1 mutation in leukemogenesis in vivo has not been fully elucidated. We report a novel mouse model that conditionally expresses the most common human NPM1 mutation (type A) in the hematopoietic compartment. In Npm1-TCTG/WT;Cre(+) mice, the NPM1 mutant localized in the cytoplasm (NPMc(+)) of bone marrow (BM) cells. The mutant mice developed no AML after 1.5-year follow-up. However, NPMc(+) expression determined a significant platelet count reduction and an expansion of the megakaryocytic compartment in the BM and spleen. Serum thrombopoietin levels overlapped in mutant vs control mice, and BM cells from Npm1-TCTG/WT;Cre(+) mice formed more megakaryocytic colonies in vitro. Moreover, we demonstrated the up-regulation of microRNAs (miRNAs; miR-10a, miR-10b, and miR-20a) inhibiting megakaryocytic differentiation along with increased expression of HOXB genes. Notably, these findings mimic those of human NPM1-mutated AML, which also exhibits a similar miRNA profile and expansion of the megakaryocytic compartment. Our mouse model provides evidence that the NPM1 mutant affects megakaryocytic development, further expanding our knowledge of the role of NPM1 mutant in leukemogenesis.
- Published
- 2013
- Full Text
- View/download PDF
28. Nucleophosmin mutations alter its nucleolar localization by impairing G-quadruplex binding at ribosomal DNA.
- Author
-
Chiarella S, De Cola A, Scaglione GL, Carletti E, Graziano V, Barcaroli D, Lo Sterzo C, Di Matteo A, Di Ilio C, Falini B, Arcovito A, De Laurenzi V, and Federici L
- Subjects
- Amino Acid Sequence, Amino Acid Substitution, Base Sequence, Binding, Competitive, Cell Line, Cell Survival drug effects, DNA, Ribosomal chemistry, DNA, Ribosomal metabolism, Humans, Kinetics, Molecular Sequence Data, Mutagenesis, Site-Directed, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Nucleophosmin, Oligonucleotides chemistry, Porphyrins chemistry, Porphyrins pharmacology, Protein Binding, Protein Structure, Tertiary, Protein Transport, Cell Nucleolus metabolism, DNA, Ribosomal genetics, G-Quadruplexes, Nuclear Proteins genetics
- Abstract
Nucleophosmin (NPM1) is an abundant nucleolar protein implicated in ribosome maturation and export, centrosome duplication and response to stress stimuli. NPM1 is the most frequently mutated gene in acute myeloid leukemia. Mutations at the C-terminal domain led to variant proteins that aberrantly and stably translocate to the cytoplasm. We have previously shown that NPM1 C-terminal domain binds with high affinity G-quadruplex DNA. Here, we investigate the structural determinants of NPM1 nucleolar localization. We show that NPM1 interacts with several G-quadruplex regions found in ribosomal DNA, both in vitro and in vivo. Furthermore, the most common leukemic NPM1 variant completely loses this activity. This is the consequence of G-quadruplex-binding domain destabilization, as mutations aimed at refolding the leukemic variant also result in rescuing the G-quadruplex-binding activity and nucleolar localization. Finally, we show that treatment of cells with a G-quadruplex selective ligand results in wild-type NPM1 dislocation from nucleoli into nucleoplasm. In conclusion, this work establishes a direct correlation between NPM1 G-quadruplex binding at rDNA and its nucleolar localization, which is impaired in the acute myeloid leukemia-associated protein variants.
- Published
- 2013
- Full Text
- View/download PDF
29. Mutational landscape of AML with normal cytogenetics: biological and clinical implications.
- Author
-
Martelli MP, Sportoletti P, Tiacci E, Martelli MF, and Falini B
- Subjects
- Biomarkers, Tumor, Cytogenetic Analysis, Genetic Heterogeneity, High-Throughput Nucleotide Sequencing, Humans, Isoenzymes genetics, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute pathology, Nucleophosmin, Prognosis, Risk Assessment, CCAAT-Enhancer-Binding Proteins genetics, Gene Expression Regulation, Leukemic, Isocitrate Dehydrogenase genetics, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Acute myeloid leukemia (AML) is a molecularly heterogeneous disease. Based on cytogenetics and FISH, AML patients are stratified into three major risk categories: favourable, intermediate and unfavourable. However, prognostic stratification and treatment decision for the intermediate risk category, that mostly comprises AML patients with normal cytogenetics (CN-AML), has been difficult due to the clinical heterogeneity and scarce knowledge of the molecular alterations underlying this large AML subgroup. During the past decade, the identification of several mutations associated with CN-AML has resulted into important advances in the AML field. In this review, we address the biological features of the main mutations associated with CN-AML and the impact of next generation sequencing studies in expanding our knowledge of the molecular landscape of CN-AML. In addition, we outline the prognostic value of mutations for risk stratification of CN-AML patients and discuss the potential of mutations discovery process for developing new molecular targeted therapies., (Copyright © 2012 Elsevier Ltd. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
30. The NPM1 wild-type OCI-AML2 and the NPM1-mutated OCI-AML3 cell lines carry DNMT3A mutations.
- Author
-
Tiacci E, Spanhol-Rosseto A, Martelli MP, Pasqualucci L, Quentmeier H, Grossmann V, Drexler HG, and Falini B
- Subjects
- Base Sequence, Cell Line, Tumor, DNA Methyltransferase 3A, Exons, Genotype, Humans, Nucleophosmin, DNA (Cytosine-5-)-Methyltransferases genetics, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics
- Published
- 2012
- Full Text
- View/download PDF
31. Acute myeloid leukemia with mutated nucleophosmin (NPM1): any hope for a targeted therapy?
- Author
-
Falini B, Gionfriddo I, Cecchetti F, Ballanti S, Pettirossi V, and Martelli MP
- Subjects
- Animals, Antineoplastic Agents pharmacology, Epigenesis, Genetic drug effects, Humans, Leukemia, Myeloid, Acute metabolism, Nuclear Proteins metabolism, Nucleophosmin, Protein Transport drug effects, Antineoplastic Agents therapeutic use, Leukemia, Myeloid, Acute drug therapy, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics
- Abstract
Acute myeloid leukemia (AML) carrying nucleophosmin (NPM1) mutations displays distinct molecular and clinical-pathological features that led to its inclusion as provisional entity in 2008 WHO classification of myeloid neoplasms. Since NPM1 mutations behave as a founder genetic lesion in AML, they could be an attractive target for therapeutic intervention. Here, we discuss the potential for developing targeted therapies for NPM1-mutated AML with focus on: (i) interfering with the abnormal traffic of the NPM1 leukemic mutant, i.e., its cytoplasmic dislocation; (ii) disrupting the nucleolar structure/function by interfering with residual wild-type nucleophosmin and other nucleolar components acting as hub proteins; and (iii) evaluating the activity of epigenetic drugs (e.g., 5-azacytidine) or agents acting on differentiation and apoptosis. As quantitative assessment of NPM1 mutated transcript copies now provides the means to measure minimal residual disease, we also discuss the potential for intervening in NPM1-mutated AML before overt hematological relapse occurs (so-called pre-emptive therapy)., (Copyright © 2011 Elsevier Ltd. All rights reserved.)
- Published
- 2011
- Full Text
- View/download PDF
32. High CD33 expression levels in acute myeloid leukemia cells carrying the nucleophosmin (NPM1) mutation.
- Author
-
De Propris MS, Raponi S, Diverio D, Milani ML, Meloni G, Falini B, Foà R, and Guarini A
- Subjects
- Adult, Aged, Aged, 80 and over, Female, Humans, Male, Middle Aged, Nucleophosmin, Sialic Acid Binding Ig-like Lectin 3, Young Adult, fms-Like Tyrosine Kinase 3 genetics, Antigens, CD metabolism, Antigens, Differentiation, Myelomonocytic metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mutation, Nuclear Proteins genetics
- Abstract
The CD33 antigen is expressed on the blast cells of most cases of acute myeloid leukemia and represents a suitable tumor-associated target antigen for antibody-based therapies. The aim of this study was to investigate the relationship between the CD33 levels quantified by mean fluorescence intensity and antibody binding capacity, and the presence/absence of NPM1 and FLT3 gene mutations in 99 newly diagnosed acute myeloid leukemia cases. The CD33 intensity evaluated as mean fluorescence intensity and antibody binding capacity was significantly higher in the NPM1-mutated acute myeloid leukemia cases compared to the NPM1-unmutated cases (P=0.0001 and P=0.0088, respectively). On the contrary, FLT3 gene mutations did not influence the levels of CD33 expression on the leukemic cells. These results establish a rational basis for the therapeutic use of anti-CD33 antibodies in NPM1-mutated acute myeloid leukemia patients.
- Published
- 2011
- Full Text
- View/download PDF
33. NPM1-mutated AML: targeting by disassembling.
- Author
-
Falini B and Martelli MP
- Subjects
- Animals, Female, Humans, Nucleophosmin, Antineoplastic Agents pharmacology, Cell Differentiation drug effects, Nuclear Proteins antagonists & inhibitors, Nuclear Proteins physiology
- Published
- 2011
- Full Text
- View/download PDF
34. Acute myeloid leukemia with mutated nucleophosmin (NPM1): is it a distinct entity?
- Author
-
Falini B, Martelli MP, Bolli N, Sportoletti P, Liso A, Tiacci E, and Haferlach T
- Subjects
- Animals, Founder Effect, Humans, Leukemia, Myeloid, Acute diagnosis, Models, Biological, Mutant Proteins genetics, Mutant Proteins metabolism, Mutant Proteins physiology, Nuclear Proteins metabolism, Nucleophosmin, Prognosis, Leukemia, Myeloid, Acute classification, Leukemia, Myeloid, Acute genetics, Mutation physiology, Nuclear Proteins genetics
- Abstract
After the discovery of NPM1-mutated acute myeloid leukemia (AML) in 2005 and its subsequent inclusion as a provisional entity in the 2008 World Health Organization classification of myeloid neoplasms, several controversial issues remained to be clarified. It was unclear whether the NPM1 mutation was a primary genetic lesion and whether additional chromosomal aberrations and multilineage dysplasia had any impact on the biologic and prognostic features of NPM1-mutated AML. Moreover, it was uncertain how to classify AML patients who were double-mutated for NPM1 and CEBPA. Recent studies have shown that: (1) the NPM1 mutant perturbs hemopoiesis in experimental models; (2) leukemic stem cells from NPM1-mutated AML patients carry the mutation; and (3) the NPM1 mutation is usually mutually exclusive of biallelic CEPBA mutations. Moreover, the biologic and clinical features of NPM1-mutated AML do not seem to be significantly influenced by concomitant chromosomal aberrations or multilineage dysplasia. Altogether, these pieces of evidence point to NPM1-mutated AML as a founder genetic event that defines a distinct leukemia entity accounting for approximately one-third of all AML.
- Published
- 2011
- Full Text
- View/download PDF
35. Nucleophosmin C-terminal leukemia-associated domain interacts with G-rich quadruplex forming DNA.
- Author
-
Federici L, Arcovito A, Scaglione GL, Scaloni F, Lo Sterzo C, Di Matteo A, Falini B, Giardina B, and Brunori M
- Subjects
- Amino Acid Sequence, DNA metabolism, Humans, Kinetics, Leukemia, Myeloid, Acute genetics, Molecular Sequence Data, Nuclear Proteins genetics, Nucleophosmin, Protein Binding, Protein Structure, Secondary, Protein Structure, Tertiary, DNA chemistry, G-Quadruplexes, Leukemia, Myeloid, Acute metabolism, Nuclear Proteins chemistry, Nuclear Proteins metabolism
- Abstract
Nucleophosmin (NPM1) is a nucleocytoplasmic shuttling phosphoprotein, mainly localized at nucleoli, that plays a key role in ribogenesis, centrosome duplication, and response to stress stimuli. Mutations at the C-terminal domain of NPM1 are the most frequent genetic lesion in acute myeloid leukemia and cause the aberrant and stable translocation of the protein in the cytoplasm. The NPM1 C-terminal domain was previously shown to bind nucleic acids. Here we further investigate the DNA binding properties of the NPM1 C-terminal domain both at the protein and nucleic acid levels; we investigate the domain boundaries and identify key residues for high affinity recognition. Furthermore, we demonstrate that the NPM1 C-terminal domain has a preference for G-quadruplex forming DNA regions and induces the formation of G-quadruplex structures in vitro. Finally we show that a specific sequence found at the SOD2 gene promoter, which was previously shown to be a target of NPM1 in vivo, is indeed folded as a G-quadruplex in vitro under physiological conditions. Our data extend considerably present knowledge on the DNA binding properties of NPM1 and suggest a general role in the transcription of genes characterized by the presence of G-quadruplex forming regions at their promoters.
- Published
- 2010
- Full Text
- View/download PDF
36. CD34+ cells from AML with mutated NPM1 harbor cytoplasmic mutated nucleophosmin and generate leukemia in immunocompromised mice.
- Author
-
Martelli MP, Pettirossi V, Thiede C, Bonifacio E, Mezzasoma F, Cecchini D, Pacini R, Tabarrini A, Ciurnelli R, Gionfriddo I, Manes N, Rossi R, Giunchi L, Oelschlägel U, Brunetti L, Gemei M, Delia M, Specchia G, Liso A, Di Ianni M, Di Raimondo F, Falzetti F, Del Vecchio L, Martelli MF, and Falini B
- Subjects
- ADP-ribosyl Cyclase 1 metabolism, Animals, Cytoplasm metabolism, Humans, Immunophenotyping, Leukemia, Myeloid, Acute metabolism, Leukemia, Myeloid, Acute pathology, Membrane Glycoproteins metabolism, Mice, Mice, Inbred NOD, Mice, SCID, Mutant Proteins metabolism, Neoplasm Transplantation, Nuclear Proteins metabolism, Nucleophosmin, Transplantation, Heterologous, Antigens, CD34 metabolism, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute immunology, Mutant Proteins genetics, Nuclear Proteins genetics
- Abstract
Acute myeloid leukemia (AML) with mutated NPM1 shows distinctive biologic and clinical features, including absent/low CD34 expression, the significance of which remains unclear. Therefore, we analyzed CD34(+) cells from 41 NPM1-mutated AML. At flow cytometry, 31 of 41 samples contained less than 10% cells showing low intensity CD34 positivity and variable expression of CD38. Mutational analysis and/or Western blotting of purified CD34(+) cells from 17 patients revealed NPM1-mutated gene and/or protein in all. Immunohistochemistry of trephine bone marrow biopsies and/or flow cytometry proved CD34(+) leukemia cells from NPM1-mutated AML had aberrant nucleophosmin expression in cytoplasm. NPM1-mutated gene and/or protein was also confirmed in a CD34(+) subfraction exhibiting the phenotype (CD34(+)/CD38(-)/CD123(+)/CD33(+)/CD90(-)) of leukemic stem cells. When transplanted into immunocompromised mice, CD34(+) cells generated a leukemia recapitulating, both morphologically and immunohistochemically (aberrant cytoplasmic nucleophosmin, CD34 negativity), the original patient's disease. These results indicate that the CD34(+) fraction in NPM1-mutated AML belongs to the leukemic clone and contains NPM1-mutated cells exhibiting properties typical of leukemia-initiating cells. CD34(-) cells from few cases (2/15) also showed significant leukemia-initiating cell potential in immunocompromised mice. This study provides further evidence that NPM1 mutation is a founder genetic lesion and has potential implications for the cell-of-origin and targeted therapy of NPM1-mutated AML.
- Published
- 2010
- Full Text
- View/download PDF
37. Rapid flow cytometric detection of aberrant cytoplasmic localization of nucleophosmin (NPMc) indicating mutant NPM1 gene in acute myeloid leukemia.
- Author
-
Oelschlaegel U, Koch S, Mohr B, Schaich M, Falini B, Ehninger G, and Thiede C
- Subjects
- Amino Acid Sequence, Base Sequence, Blast Crisis diagnosis, Blast Crisis genetics, Bone Marrow metabolism, Bone Marrow pathology, Case-Control Studies, Humans, Leukemia, Myeloid, Acute diagnosis, Molecular Sequence Data, Multicenter Studies as Topic, Nucleophosmin, Prognosis, Protein Transport, Sequence Homology, Amino Acid, Sequence Homology, Nucleic Acid, Cytoplasm metabolism, Cytoplasm pathology, Flow Cytometry, Leukemia, Myeloid, Acute genetics, Mutation genetics, Nuclear Proteins genetics
- Published
- 2010
- Full Text
- View/download PDF
38. NPM1 deletion is associated with gross chromosomal rearrangements in leukemia.
- Author
-
La Starza R, Matteucci C, Gorello P, Brandimarte L, Pierini V, Crescenzi B, Nofrini V, Rosati R, Gottardi E, Saglio G, Santucci A, Berchicci L, Arcioni F, Falini B, Martelli MF, Sambani C, Aventin A, and Mecucci C
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Base Sequence, Child, Chromosome Deletion, Chromosomes, Human, Pair 5 genetics, Chromosomes, Human, Pair 5 metabolism, Female, Humans, Leukemia, Myeloid metabolism, Male, Middle Aged, Molecular Sequence Data, Nuclear Proteins metabolism, Nucleophosmin, Young Adult, Gene Deletion, Leukemia, Myeloid genetics, Nuclear Proteins genetics, Translocation, Genetic
- Abstract
Background: NPM1 gene at chromosome 5q35 is involved in recurrent translocations in leukemia and lymphoma. It also undergoes mutations in 60% of adult acute myeloid leukemia (AML) cases with normal karyotype. The incidence and significance of NPM1 deletion in human leukemia have not been elucidated., Methodology and Principal Findings: Bone marrow samples from 145 patients with myelodysplastic syndromes (MDS) and AML were included in this study. Cytogenetically 43 cases had isolated 5q-, 84 cases had 5q- plus other changes and 18 cases had complex karyotype without 5q deletion. FISH and direct sequencing investigated the NPM1 gene. NPM1 deletion was an uncommon event in the "5q- syndrome" but occurred in over 40% of cases with high risk MDS/AML with complex karyotypes and 5q loss. It originated from large 5q chromosome deletions. Simultaneous exon 12 mutations were never found. NPM1 gene status was related to the pattern of complex cytogenetic aberrations. NPM1 haploinsufficiency was significantly associated with monosomies (p<0.001) and gross chromosomal rearrangements, i.e., markers, rings, and double minutes (p<0.001), while NPM1 disomy was associated with structural changes (p=0.013). Interestingly, in complex karyotypes with 5q- TP53 deletion and/or mutations are not specifically associated with NPM1 deletion., Conclusions and Significance: NPM1/5q35 deletion is a consistent event in MDS/AML with a 5q-/-5 in complex karyotypes. NPM1 deletion and NPM1 exon 12 mutations appear to be mutually exclusive and are associated with two distinct cytogenetic subsets of MDS and AML.
- Published
- 2010
- Full Text
- View/download PDF
39. Multilineage dysplasia has no impact on biologic, clinicopathologic, and prognostic features of AML with mutated nucleophosmin (NPM1).
- Author
-
Falini B, Macijewski K, Weiss T, Bacher U, Schnittger S, Kern W, Kohlmann A, Klein HU, Vignetti M, Piciocchi A, Fazi P, Martelli MP, Vitale A, Pileri S, Miesner M, Santucci A, Haferlach C, Mandelli F, and Haferlach T
- Subjects
- Female, Gene Expression Profiling, Gene Expression Regulation, Leukemic, Humans, Karyotyping, Leukemia, Myeloid, Acute pathology, Male, Myelodysplastic Syndromes pathology, Nucleophosmin, Oligonucleotide Array Sequence Analysis, Prognosis, Survival Rate, fms-Like Tyrosine Kinase 3 genetics, Biomarkers, Tumor genetics, Cell Lineage, Leukemia, Myeloid, Acute genetics, Mutation genetics, Myelodysplastic Syndromes genetics, Nuclear Proteins genetics
- Abstract
NPM1-mutated acute myeloid leukemia (AML) is a provisional entity in the 2008 World Health Organization (WHO) classification of myeloid neoplasms. The significance of multilineage dysplasia (MLD) in NPM1-mutated AML is unclear. Thus, in the 2008 WHO classification, NPM1-mutated AML with MLD is classified as AML with myelodysplasia (MD)-related changes (MRCs). We evaluated morphologically 318 NPM1-mutated AML patients and found MLD in 23.3%. Except for a male predominance and a lower fms-related tyrosine kinase 3-internal tandem duplication (FLT3-ITD) incidence in the MLD(+) group, no differences were observed in age, sex, cytogenetics, and FLT3--tyrosine kinase domain between NPM1-mutated AML with and without MLD. NPM1-mutated AML with and without MLD showed overlapping immunophenotype (CD34 negativity) and gene expression profile (CD34 down-regulation, HOX genes up-regulation). Moreover, overall and event-free survival did not differ among NPM1-mutated AML patients independently of whether they were MLD(+) or MLD(-), the NPM1-mutated/FLT3-ITD negative genotype showing the better prognosis. Lack of MLD impact on survival was confirmed by multivariate analysis that highlighted FLT3-ITD as the only significant prognostic parameter in NPM1-mutated AML. Our findings indicate that NPM1 mutations rather than MLD dictate the distinctive features of NPM1-mutated AML. Thus, irrespective of MLD, NPM1-mutated AML represents one disease entity clearly distinct from AML with MRCs.
- Published
- 2010
- Full Text
- View/download PDF
40. Expression of the cytoplasmic NPM1 mutant (NPMc+) causes the expansion of hematopoietic cells in zebrafish.
- Author
-
Bolli N, Payne EM, Grabher C, Lee JS, Johnston AB, Falini B, Kanki JP, and Look AT
- Subjects
- Animals, Apoptosis genetics, Base Sequence, Blotting, Western, Cell Separation, Cytoplasm metabolism, Embryo, Nonmammalian, Flow Cytometry, Fluorescent Antibody Technique, Hematopoietic Stem Cells physiology, Humans, Immunoprecipitation, Leukemia, Myeloid, Acute metabolism, Molecular Sequence Data, Mutation, Nuclear Proteins metabolism, Nucleophosmin, Reverse Transcriptase Polymerase Chain Reaction, Sequence Homology, Nucleic Acid, Zebrafish, Hematopoiesis genetics, Leukemia, Myeloid, Acute genetics, Myeloid Cells physiology, Nuclear Proteins genetics
- Abstract
Mutations in the human nucleophosmin (NPM1) gene are the most frequent genetic alteration in adult acute myeloid leukemias (AMLs) and result in aberrant cytoplasmic translocation of this nucleolar phosphoprotein (NPMc+). However, underlying mechanisms leading to leukemogenesis remain unknown. To address this issue, we took advantage of the zebrafish model organism, which expresses 2 genes orthologous to human NPM1, referred to as npm1a and npm1b. Both genes are ubiquitously expressed, and their knockdown produces a reduction in myeloid cell numbers that is specifically rescued by NPM1 expression. In zebrafish, wild-type human NPM1 is nucleolar while NPMc+ is cytoplasmic, as in human AML, and both interact with endogenous zebrafish Npm1a and Npm1b. Forced NPMc+ expression in zebrafish causes an increase in pu.1(+) primitive early myeloid cells. A more marked perturbation of myelopoiesis occurs in p53(m/m) embryos expressing NPMc+, where mpx(+) and csf1r(+) cell numbers are also expanded. Importantly, NPMc+ expression results in increased numbers of definitive hematopoietic cells, including erythromyeloid progenitors in the posterior blood island and c-myb/cd41(+) cells in the ventral wall of the aorta. These results are likely to be relevant to human NPMc+ AML, where the observed NPMc+ multilineage expression pattern implies transformation of a multipotent stem or progenitor cell.
- Published
- 2010
- Full Text
- View/download PDF
41. Molecular and alternative methods for diagnosis of acute myeloid leukemia with mutated NPM1: flexibility may help.
- Author
-
Falini B, Martelli MP, Pileri SA, and Mecucci C
- Subjects
- Humans, Leukemia, Myeloid, Acute blood, Nucleophosmin, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics
- Published
- 2010
- Full Text
- View/download PDF
42. Acute myeloid leukemia with mutated nucleophosmin (NPM1): molecular, pathological, and clinical features.
- Author
-
Falini B
- Subjects
- Adult, Centrosome metabolism, Child, Exons genetics, Forecasting, Humans, Leukemia, Myeloid, Acute classification, Leukemia, Myeloid, Acute diagnosis, Leukemia, Myeloid, Acute pathology, Nuclear Proteins physiology, Nucleophosmin, Phosphorylation, Protein Processing, Post-Translational, Protein Transport genetics, Protein Transport physiology, Ribosomes metabolism, Tumor Suppressor Proteins metabolism, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics
- Abstract
The NPM1 gene encodes for nucleophosmin, a nucleolus-located shuttling protein that is involved in multiple cell functions, including regulation of ribosome biogenesis, control of centrosome duplication and preservation of ARF tumor suppressor integrity. The NPM1 gene is specifically mutated in about 30% acute myeloid leukemia (AML) but not in other human neoplasms. Mutations cause crucial changes at the C-terminus of the NPM1 protein that are responsible for the aberrant nuclear export and accumulation of NPM1 mutants in the cytoplasm of leukemic cells. Diagnosis of AML with mutated NPM1 can be done using molecular techniques, immunohistochemistry (looking at cytoplasmic dislocation of nucleophosmin that is predictive of NPM1 mutations) and Western blotting with antibodies specifically directed against NPM1 mutants. Because of its distinctive molecular, pathological, immunophenotypic and prognostic features, AML with mutated NPM1 (synonym: NPMc+ AML) has been included, as a new provisional entity, in the 2008 World Health Organization (WHO) classification of myeloid neoplasms.
- Published
- 2010
- Full Text
- View/download PDF
43. Cuplike nuclei (prominent nuclear invaginations) in acute myeloid leukemia are highly associated with FLT3 internal tandem duplication and NPM1 mutation.
- Author
-
Chen W, Konoplev S, Medeiros LJ, Koeppen H, Leventaki V, Vadhan-Raj S, Jones D, Kantarjian HM, Falini B, and Bueso-Ramos CE
- Subjects
- Adolescent, Adult, Aged, Aged, 80 and over, Cell Nucleus genetics, Female, Gene Duplication, Humans, Immunophenotyping, Male, Middle Aged, Mutation, Nucleophosmin, Cell Nucleus pathology, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute pathology, Nuclear Proteins genetics, fms-Like Tyrosine Kinase 3 genetics
- Abstract
Background: A small subset of patients with acute myeloid leukemia (AML) have cuplike nuclei. Other investigators have demonstrated that these neoplasms have distinctive clinicopathologic and molecular features., Methods: The authors searched for patients who had AML with cuplike nuclei at their institution over a 10-year interval. A strict definition for cuplike nuclei was used: >or=10% blasts with nuclear invaginations in >or=25% of the nuclear area. The relevant data were reviewed, and the results were compared with a control group of patients who had AML without cuplike nuclei., Results: In total, 22 patients who had AML with cuplike nuclei were identified and were classified as AML without maturation (French-American-British classification M1) (AML M1). Compared with the control group (AML M1), patients who had AML with cuplike nuclei were associated significantly with fms-like tyrosine kinase 3 (FLT3)-internal tandem duplication (ITD) (86% vs 38%, respectively; P = .002); nucleophosmin 1 (NPM1) mutations (86% vs 19%; P < .0001); both mutations (77% vs 14%; P < .0001); normal karyotype (86% vs 40%; P = .003); bone marrow blast count (90% vs 84%; P = .016); myeloperoxidase positivity (95% vs 30% blasts; P = .001); higher D-dimer levels (>5000 ng/mL vs 569 ng/mL; P = .001); and the absence of CD7 (91% vs 52%; P = .007), CD34 (82% vs 5%; P < .0001), and human leukocyte antigen, D-related (59% vs 10%; P = .001). There were no differences in age, sex, or peripheral blood counts. The positive predictive value of recognizing AML with cuplike nuclei for FLT3-ITD, NPM1, and both mutations was 81%, 86%, and 77%, respectively., Conclusions: Cuplike nuclei in AML were highly associated with the presence of NPM1 and FLT3-ITD mutations and with several clinicopathologic and immunophenotypic features. Recognition of the distinctive morphologic features of AML with cuplike nuclei may be helpful in streamlining the workup of these neoplasms., ((c) 2009 American Cancer Society.)
- Published
- 2009
- Full Text
- View/download PDF
44. Acute myeloid leukemia with mutated NPM1: diagnosis, prognosis and therapeutic perspectives.
- Author
-
Falini B, Sportoletti P, and Martelli MP
- Subjects
- Acute Disease, Base Sequence, Genetic Predisposition to Disease, Humans, Leukemia, Myeloid diagnosis, Leukemia, Myeloid therapy, Molecular Sequence Data, Nucleophosmin, Prognosis, Leukemia, Myeloid genetics, Mutation, Nuclear Proteins genetics
- Abstract
Purpose of Review: Nucleophosmin (NPM1) gene mutations, which cause aberrant cytoplasmic expression of nucleophosmin (NPMc+), are the most frequent genetic alteration in acute myeloid leukemia (AML), being found in about 30% cases. The present review summarizes recent advances in the biology, diagnosis, prognosis and therapy of NPM1-mutated AML., Recent Findings: Diagnostic criteria of NPM1-mutated AML are discussed in the light of its recent inclusion in the 2008 WHO classification of myeloid neoplasms. We also outline the most recent findings on prognosis and monitoring of minimal residual disease in NPM1-mutated AML and their implications for therapeutic decisions. Moreover, new insights are presented into the molecular mechanisms underlying perturbed nucleophosmin traffic in NPM1-mutated AML, which provides the rationale for the development of targeted therapies., Summary: AML with mutated NPM1 is a leukemia entity with distinct molecular, pathological, and prognostic features.
- Published
- 2009
- Full Text
- View/download PDF
45. AML with mutated NPM1 carrying a normal or aberrant karyotype show overlapping biologic, pathologic, immunophenotypic, and prognostic features.
- Author
-
Haferlach C, Mecucci C, Schnittger S, Kohlmann A, Mancini M, Cuneo A, Testoni N, Rege-Cambrin G, Santucci A, Vignetti M, Fazi P, Martelli MP, Haferlach T, and Falini B
- Subjects
- Gene Expression Profiling, Gene Expression Regulation, Leukemic, Homeodomain Proteins genetics, Homeodomain Proteins metabolism, Humans, Immunophenotyping, Karyotyping, Leukemia, Myeloid, Acute pathology, Nucleophosmin, Oligonucleotide Array Sequence Analysis, Prognosis, RNA, Messenger genetics, RNA, Messenger metabolism, Reverse Transcriptase Polymerase Chain Reaction, fms-Like Tyrosine Kinase 3 genetics, Antigens, CD34 metabolism, Chromosome Aberrations, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute immunology, Mutation genetics, Nuclear Proteins genetics
- Abstract
Acute myeloid leukemia (AML) with mutated NPM1 usually carries normal karyotype (NK), but it may harbor chromosomal aberrations whose significance remains unclear. We addressed this question in 631 AML patients with mutated/cytoplasmic NPM1. An abnormal karyotype (AK) was present in 93 of 631 cases (14.7%), the most frequent abnormalities being +8, +4, -Y, del(9q), +21. Chromosome aberrations in NPM1-mutated AML were similar to, but occurred less frequently than additional chromosome changes found in other AML with recurrent cytogenetic abnormalities according to WHO classification. Four of the 31 NPM1-mutated AML patients karyotyped at different time points had NK at diagnosis but AK at relapse: del(9q) (n = 2), t(2;11) (n = 1), inv(12) (n = 1). NPM1-mutated AML with NK or AK showed overlapping morphologic, immunophenotypic (CD34 negativity), and gene expression profile (down-regulation of CD34 and up-regulation of HOX genes). No difference in survival was observed among NPM1-mutated AML patients independently of whether they carried a NK or an AK, the NPM1-mutated/FLT3-ITD negative cases showing the better prognosis. Findings in our patients point to chromosomal aberrations as secondary events, reinforce the concept that NPM1 mutation is a founder genetic lesion, and indicate that NPM1-mutated AML should be clinically handled as one entity, irrespective of the karyotype.
- Published
- 2009
- Full Text
- View/download PDF
46. Altered nucleophosmin transport in acute myeloid leukaemia with mutated NPM1: molecular basis and clinical implications.
- Author
-
Falini B, Bolli N, Liso A, Martelli MP, Mannucci R, Pileri S, and Nicoletti I
- Subjects
- Humans, Nucleophosmin, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Mutation genetics, Nuclear Proteins genetics, Nuclear Proteins metabolism
- Abstract
Nucleophosmin (NPM1) is a highly conserved nucleo-cytoplasmic shuttling protein that shows a restricted nucleolar localization. Mutations of NPM1 gene leading to aberrant cytoplasmic dislocation of nucleophosmin (NPMc+) occurs in about one third of acute myeloid leukaemia (AML) patients that exhibit distinctive biological and clinical features. We discuss the latest advances in the molecular basis of nucleophosmin traffic under physiological conditions, describe the molecular abnormalities underlying altered transport of nucleophosmin in NPM1-mutated AML and present evidences supporting the view that cytoplasmic nucleophosmin is a critical event for leukaemogenesis. We then outline how a highly specific immunohistochemical assay can be exploited to diagnose NPM1-mutated AML and myeloid sarcoma in paraffin-embedded samples by looking at aberrant nucleophosmin accumulation in cytoplasm of leukaemic cells. This procedure is also suitable for detection of haemopoietic multilineage involvement in bone marrow trephines. Moreover, use of immunohistochemistry as surrogate for molecular analysis can serve as first-line screening in AML and should facilitate implementation of the 2008 World Health Organization classification of myeloid neoplasms that now incorporates AML with mutated NPM1 (synonym: NPMc+ AML) as a new provisional entity. Finally, we discuss the future therapeutic perspectives aimed at reversing the altered nucleophosmin transport in AML with mutated NPM1.
- Published
- 2009
- Full Text
- View/download PDF
47. Minimal residual disease levels assessed by NPM1 mutation-specific RQ-PCR provide important prognostic information in AML.
- Author
-
Schnittger S, Kern W, Tschulik C, Weiss T, Dicker F, Falini B, Haferlach C, and Haferlach T
- Subjects
- Adult, Age Factors, Aged, Antigens, CD34 genetics, Antigens, CD34 metabolism, Female, Follow-Up Studies, Humans, Leukemia, Myeloid, Acute blood, Leukemia, Myeloid, Acute therapy, Leukocyte Count, Male, Middle Aged, Neoplasm, Residual, Nuclear Proteins metabolism, Nucleophosmin, Prognosis, Proto-Oncogene Proteins c-abl genetics, Proto-Oncogene Proteins c-abl metabolism, Recurrence, Stem Cell Transplantation, Transplantation, Homologous, fms-Like Tyrosine Kinase 3 genetics, fms-Like Tyrosine Kinase 3 metabolism, Leukemia, Myeloid, Acute genetics, Mutation, Nuclear Proteins genetics, Polymerase Chain Reaction
- Abstract
Nucleophosmin (NPM1)-mutated acute myeloid leukemia (AML), which is recognized as a provisional entity in the World Health Organization 2008 classification of myeloid neoplasms, accounts for 30% of AML. We analyzed 1227 diagnostic and follow-up samples in 252 NPM1-mutated AML patients with 17 different NPM1 mutation-specific real-time quantitative polymerase chain reaction (RQ-PCR) assays. Paired diagnostic/relapse samples of 84 patients revealed stable NPM1 mutations in all cases, suggesting that they are pathogenetically early events and thus applicable for minimal residual disease detection. A total of 47 relapses were predictable because of an NPM1 mutation level (%NPM1/ABL1) increase of at least 1 log or in 15 cases because of NPM1 mutation levels not decreasing less than 3 log ranges. A high prognostic value of NPM1 levels was shown for 4 different intervals after therapy was initiated. Furthermore, thresholds of 0.1 and 0.01%NPM1/ABL1 during/after treatment discriminated between prognostic subgroups. Univariate analyses, including age, white blood cell count, blast count, CD34 positivity, FLT3 mutations status, FAB type, karyotype, NPM1 mutation type, and pretreatment NPM1 mutational level, showed that, besides NPM1 mutation level, only age and FLT3-LM mutation status were prognostically significant for EFS. Multivariate analysis, including age, FLT3-LM status, and NPM1 mutation level at different time points, demonstrated that NPM1 level was the most relevant prognostic factor during first-line treatment. Similar results were obtained in patients undergoing second-line chemotherapy or allogeneic stem cell transplantation.
- Published
- 2009
- Full Text
- View/download PDF
48. Folding mechanism of the C-terminal domain of nucleophosmin: residual structure in the denatured state and its pathophysiological significance.
- Author
-
Scaloni F, Gianni S, Federici L, Falini B, and Brunori M
- Subjects
- Humans, In Vitro Techniques, Kinetics, Leukemia, Myeloid, Acute genetics, Leukemia, Myeloid, Acute metabolism, Models, Molecular, Mutation, Nuclear Proteins genetics, Nuclear Proteins physiology, Nucleophosmin, Protein Denaturation, Protein Folding, Protein Structure, Tertiary, Recombinant Proteins chemistry, Recombinant Proteins genetics, Recombinant Proteins metabolism, Thermodynamics, Nuclear Proteins chemistry
- Abstract
Nucleophosmin (NPM1) is a ubiquitously expressed protein and is one of the most abundant proteins found in the nucleolus. Naturally occurring mutations in the C-terminal domain of nucleophosmin (Cter-NPM1) are found in approximately 30% of patients with acute myeloid leukemia (AML). These mutations cause changes at the C terminus of NPM1 that lead to denaturation of the protein, a critical factor in determining aberrant translocation of NPM1 to the cytosol. Hence, this protein system represents an ideal candidate to investigate the relations between folding and unfolding and disease. Here we report the characterization of the folding and unfolding kinetics of Cter-NPM1. Data reveal that this small helical domain folds via a compact denatured state, displaying a malleable residual structure. Moreover, analysis of folding rate constants measured under different experimental conditions suggests that the existence of a preorganized structure in the denatured state accelerates folding, implying a native-like residual structure. Because a major feature of Cter-NPM1 mutants responsible for AML is a reduction in stability of the protein and thus prevalence of a denatured state even under physiological conditions, our findings may pave the way to further studies with the aim of designing chemicals capable of interacting with the "pathological" mutants to stabilize the native conformation.
- Published
- 2009
- Full Text
- View/download PDF
49. Cytoplasmic mutated nucleophosmin (NPM1) in blast crisis of chronic myeloid leukaemia.
- Author
-
Piccaluga PP, Sabattini E, Bacci F, Agostinelli C, Righi S, Salmi F, Testoni N, Paolini S, Castagnetti F, Martinelli G, Falini B, and Pileri SA
- Subjects
- Aged, Female, Flow Cytometry, Humans, Immunophenotyping, Leukemia, Myelogenous, Chronic, BCR-ABL Positive genetics, Nucleophosmin, Nucleoplasmins, Blast Crisis, Cytoplasm metabolism, Leukemia, Myelogenous, Chronic, BCR-ABL Positive pathology, Molecular Chaperones genetics, Mutation, Nuclear Proteins genetics, Phosphoproteins genetics
- Published
- 2009
- Full Text
- View/download PDF
50. A dose-dependent tug of war involving the NPM1 leukaemic mutant, nucleophosmin, and ARF.
- Author
-
Bolli N, De Marco MF, Martelli MP, Bigerna B, Pucciarini A, Rossi R, Mannucci R, Manes N, Pettirossi V, Pileri SA, Nicoletti I, and Falini B
- Subjects
- Active Transport, Cell Nucleus genetics, Acute Disease, Animals, Cell Nucleolus metabolism, Cell Transformation, Neoplastic genetics, Cytoplasm metabolism, Dimerization, Drug Delivery Systems, Humans, Leukemia, Myeloid metabolism, Mice, NIH 3T3 Cells metabolism, Neoplasm Proteins chemistry, Neoplasm Proteins metabolism, Nuclear Export Signals physiology, Nuclear Proteins chemistry, Nuclear Proteins metabolism, Nucleophosmin, Recombinant Fusion Proteins chemistry, Recombinant Fusion Proteins metabolism, Structure-Activity Relationship, Transfection, Tumor Suppressor Protein p14ARF metabolism, Active Transport, Cell Nucleus physiology, Leukemia, Myeloid genetics, Neoplasm Proteins genetics, Nuclear Export Signals genetics, Nuclear Proteins genetics, Protein Interaction Mapping, Tumor Suppressor Protein p14ARF chemistry
- Abstract
In acute myeloid leukaemia (AML), nucleophosmin-1 (NPM1) mutations create a nuclear export signal (NES) motif and disrupt tryptophans at NPM1 C-terminus, leading to nucleophosmin accumulation in leukaemic cell cytoplasm. We investigated how nucleophosmin NES motifs (two physiological and one created by the mutation) regulate traffic and interaction of mutated NPM1, NPM1wt and p14(ARF). Nucleophosmin export into cytoplasm was maximum when the protein contained all three NES motifs, as naturally occurs in NPM1-mutated AML. The two physiological NES motifs mediated NPM1 homo/heterodimerization, influencing subcellular distribution of NPM1wt, mutated NPM1 and p14(ARF) in a 'dose-dependent tug of war' fashion. In transfected cells, excess doses of mutant NPM1 relocated completely NPM1wt (and p14(ARF)) from the nucleoli to the cytoplasm. This distribution pattern was also observed in a proportion of NPM1-mutated AML patients. In transfected cells, excess of NPM1wt (and p14(ARF)) relocated NPM1 mutant from the cytoplasm to the nucleoli. Notably, this distribution pattern was not observed in AML patients where the mutant was consistently cytoplasmic restricted. These findings reinforce the concept that NPM1 mutants are naturally selected for most efficient cytoplasmic export, pointing to this event as critical for leukaemogenesis. Moreover, they provide a rationale basis for designing small molecules acting at the interface between mutated NPM1 and other interacting proteins.
- Published
- 2009
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.