Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Leukemia ; 36(10): 2351-2367, 2022 10.
Article in English | MEDLINE | ID: mdl-36008542

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.


Subject(s)
Leukemia, Myeloid, Acute , Nuclear Proteins , Adult , Dactinomycin/therapeutic use , Histones/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/pathology , Mutation , Nuclear Proteins/metabolism , Nucleophosmin , Proto-Oncogene Proteins c-bcl-2/metabolism , RNA , Receptors, Antigen, T-Cell/genetics
2.
Biomolecules ; 12(7)2022 06 28.
Article in English | MEDLINE | ID: mdl-35883460

ABSTRACT

The administration of combinations of drugs is a method widely used in the treatment of different pathologies as it can lead to an increase in the therapeutic effect and a reduction in the dose compared to the administration of single drugs. For these reasons, it is of interest to study combinations of drugs and to determine whether a specific combination has a synergistic, antagonistic or additive effect. Various mathematical models have been developed, which use different methods to evaluate the synergy of a combination of drugs. We have developed an open access and easy to use app that allows different models to be explored and the most fitting to be chosen for the specific experimental data: SiCoDEA (Single and Combined Drug Effect Analysis). Despite the existence of other tools for drug combination analysis, SiCoDEA remains the most complete and flexible since it offers options such as outlier removal or the ability to choose between different models for analysis. SiCoDEA is an easy to use tool for analyzing drug combination data and to have a view of the various steps and offer different results based on the model chosen.


Subject(s)
Mobile Applications , Drug Combinations , Drug Synergism , Pharmaceutical Preparations
3.
Blood ; 138(25): 2696-2701, 2021 12 23.
Article in English | MEDLINE | ID: mdl-34343258

ABSTRACT

Nucleophosmin (NPM1) mutations in acute myeloid leukemia (AML) affect exon 12, but also sporadically affect exons 9 and 11, causing changes at the protein C-terminal end (tryptophan loss, nuclear export signal [NES] motif creation) that lead to aberrant cytoplasmic NPM1 (NPM1c+), detectable by immunohistochemistry. Combining immunohistochemistry and molecular analyses in 929 patients with AML, we found non-exon 12 NPM1 mutations in 5 (1.3%) of 387 NPM1c+ cases. Besides mutations in exons 9 (n = 1) and 11 (n = 1), novel exon 5 mutations were discovered (n = 3). Another exon 5 mutation was identified in an additional 141 patients with AML selected for wild-type NPM1 exon 12. Three NPM1 rearrangements (NPM1/RPP30, NPM1/SETBP1, NPM1/CCDC28A) were detected and characterized among 13 979 AML samples screened by cytogenetic/fluorescence in situ hybridization and RNA sequencing. Functional studies demonstrated that in AML cases, new NPM1 proteins harbored an efficient extra NES, either newly created or already present in the fusion partner, ensuring its cytoplasmic accumulation. Our findings support NPM1 cytoplasmic relocation as critical for leukemogenesis and reinforce the role of immunohistochemistry in predicting AML-associated NPM1 genetic lesions. This study highlights the need to develop new assays for molecular diagnosis and monitoring of NPM1-mutated AML.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Nucleophosmin/genetics , Adult , Exons , Female , Gene Fusion , Gene Rearrangement , Humans , Male , Middle Aged
4.
Leukemia ; 35(9): 2552-2562, 2021 09.
Article in English | MEDLINE | ID: mdl-33654209

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.


Subject(s)
Cell Nucleolus/drug effects , Dactinomycin/therapeutic use , Drug Resistance, Neoplasm , Leukemia, Myeloid, Acute/drug therapy , Mutation , Neoplasm Recurrence, Local/drug therapy , Nuclear Proteins/genetics , 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
5.
Cancers (Basel) ; 13(3)2021 Jan 28.
Article in English | MEDLINE | ID: mdl-33525388

ABSTRACT

NPM1-mutated (NPM1mut) acute myeloid leukemia (AML) comprises about 30% of newly diagnosed AML in adults. Despite notable advances in the treatment of this frequent AML subtype, about 50% of NPM1mut AML patients treated with conventional treatment die due to disease progression. CD123 has been identified as potential target for immunotherapy in AML, and several anti-CD123 therapeutic approaches have been developed for AML resistant to conventional therapies. As this antigen has been previously reported to be expressed by NPM1mut cells, we performed a deep flow cytometry analysis of CD123 expression in a large cohort of NPM1mut and wild-type samples, examining the whole blastic population, as well as CD34+CD38- leukemic cells. We demonstrate that CD123 is highly expressed on NPM1mut cells, with particularly high expression levels showed by CD34+CD38- leukemic cells. Additionally, CD123 expression was further enhanced by FLT3 mutations, which frequently co-occur with NPM1 mutations. Our results identify NPM1-mutated and particularly NPM1/FLT3 double-mutated AML as disease subsets that may benefit from anti-CD123 targeted therapies.

6.
Cancer Cell ; 34(3): 499-512.e9, 2018 09 10.
Article in English | MEDLINE | ID: mdl-30205049

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.


Subject(s)
Gene Expression Regulation, Leukemic , Homeodomain Proteins/metabolism , Leukemia, Myeloid, Acute/genetics , Nuclear Proteins/genetics , 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
7.
Blood ; 125(22): 3455-65, 2015 May 28.
Article in English | MEDLINE | ID: mdl-25795919

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.


Subject(s)
Apoptosis/drug effects , Arsenicals/pharmacology , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Oxides/pharmacology , Tretinoin/pharmacology , 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
8.
Blood ; 121(17): 3447-58, 2013 Apr 25.
Article in English | MEDLINE | ID: mdl-23435463

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.


Subject(s)
Disease Models, Animal , Integrases/metabolism , Leukemia, Myeloid, Acute/etiology , Megakaryocytes/pathology , Mutation/genetics , Nuclear Proteins/genetics , Thrombopoiesis/genetics , 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
9.
Blood Rev ; 25(6): 247-54, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21724308

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).


Subject(s)
Antineoplastic Agents/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Mutation , Nuclear Proteins/genetics , Animals , Antineoplastic Agents/pharmacology , Epigenesis, Genetic/drug effects , Humans , Leukemia, Myeloid, Acute/metabolism , Nuclear Proteins/metabolism , Nucleophosmin , Protein Transport/drug effects
10.
Blood ; 116(19): 3907-22, 2010 Nov 11.
Article in English | MEDLINE | ID: mdl-20634376

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.


Subject(s)
Antigens, CD34/metabolism , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/immunology , Mutant Proteins/genetics , Nuclear Proteins/genetics , 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
11.
Cancer Lett ; 246(1-2): 274-81, 2007 Feb 08.
Article in English | MEDLINE | ID: mdl-16650526

ABSTRACT

Human Papillomavirus type 77 is a skin type found in non-melanoma skin cancers of immuno-compromised individuals. Although, the HPV77 E6 oncoprotein has been well studied, nothing is known about E7. Studies on mucosal HPV types (e.g. HPV16) showed that E7 deregulates the cell cycle by binding to and promoting degradation of retinoblastoma protein (pRb). Here, we characterized the impact of HPV77 E7 on the cell cycle. We observed that HPV77 E7 associated with pRb with a lower affinity than HPV16 E7, promoting weakly its degradation. Although, HPV16 E7 led to cellular proliferation and accumulation of the cell cycle inhibitor p16(INK4a), both events were not clearly observed in HPV77 E7 cells. Together, these data indicate that HPV77 E7 does not efficiently deregulate the cell cycle, in contrast to several E7s of mucosal HPV types.


Subject(s)
Cell Cycle/physiology , Papillomavirus E7 Proteins/metabolism , Animals , Cell Cycle/genetics , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p16/genetics , Cyclin-Dependent Kinase Inhibitor p16/metabolism , Gene Expression , Genetic Vectors/genetics , Humans , Immunoblotting , Mice , NIH 3T3 Cells , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Protein Binding , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Transfection
SELECTION OF CITATIONS
SEARCH DETAIL
...