Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cytometry A ; 103(10): 818-829, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37338802

ABSTRACT

Although most modern techniques and analysis methods in multiparameter flow cytometry (MFC) allow for increased dimensionality for the characterization and quantification of cell populations, most MFC applications depend on flow cytometers measuring relatively small (<16) numbers of parameters. When more markers than the available parameters need to be acquired, these are commonly distributed over multiple independent measurements that include a backbone of common markers. Several methods have been proposed to impute values for combinations of markers that were not measured simultaneously. These imputation methods are frequently used without proper validation and knowledge of their effects on data analysis. We evaluated the performance of existing imputation software (Infinicyt, CyTOFmerge, CytoBackBone, and cyCombine) in approximating known measured expression data in terms of similarity in visual appearance, cell expression, and gating in different datasets by splitting MFC samples into separate measurements with partially overlapping markers and re-calculating missing marker expression. Out of the assessed packages, CyTOFmerge showed the most accurate approximation of the known expression in terms of similar expression values and concordance with manual gating, with a mean F-score between 0.53 and 0.87 when retrieving cell populations in different datasets. Performance remained inadequate for all methods, with only limited similarity at the cell level. In conclusion, the use of imputed MFC data should take such limitations into account and include independent validation of results to justify conclusions.

2.
Blood ; 137(22): 3008-3009, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34081123
3.
Drug Resist Updat ; 53: 100730, 2020 12.
Article in English | MEDLINE | ID: mdl-33096284

ABSTRACT

New treatment options of acute myeloid leukemia (AML) are rapidly emerging. Pre-clinical models such as ex vivo cultures are extensively used towards the development of novel drugs and to study synergistic drug combinations, as well as to discover biomarkers for both drug response and anti-cancer drug resistance. Although these approaches empower efficient investigation of multiple drugs in a multitude of primary AML samples, their translational value and reproducibility are hampered by the lack of standardized methodologies and by culture system-specific behavior of AML cells and chemotherapeutic drugs. Moreover, distinct research questions require specific methods which rely on specific technical knowledge and skills. To address these aspects, we herein review commonly used culture techniques in light of diverse research questions. In addition, culture-dependent effects on drug resistance towards commonly used drugs in the treatment of AML are summarized including several pitfalls that may arise because of culture technique artifacts. The primary aim of the current review is to provide practical guidelines for ex vivo primary AML culture experimental design.


Subject(s)
Antineoplastic Agents/pharmacology , Leukemia, Myeloid, Acute/drug therapy , Primary Cell Culture/methods , Research Design/standards , Antineoplastic Agents/therapeutic use , Bone Marrow/pathology , Cryopreservation , Culture Media/chemistry , Culture Media/standards , Drug Resistance, Neoplasm , Drug Screening Assays, Antitumor/instrumentation , Drug Screening Assays, Antitumor/methods , Drug Screening Assays, Antitumor/standards , Guidelines as Topic , Humans , Leukemia, Myeloid, Acute/blood , Leukemia, Myeloid, Acute/pathology , Primary Cell Culture/instrumentation , Primary Cell Culture/standards , Reproducibility of Results , Tumor Cells, Cultured
5.
Leukemia ; 34(7): 1751-1759, 2020 07.
Article in English | MEDLINE | ID: mdl-32020044

ABSTRACT

More effective treatment modalities are urgently needed in patients with acute myeloid leukemia (AML) of older age. We hypothesized that adding lenalidomide to intensive standard chemotherapy might improve their outcome. After establishing a safe lenalidomide, dose elderly patients with AML were randomly assigned in this randomized Phase 2 study (n = 222) to receive standard chemotherapy ("3 + 7") with or without lenalidomide at a dose of 20 mg/day 1-21. In the second cycle, patients received cytarabine 1000 mg/m2 twice daily on days 1-6 with or without lenalidomide (20 mg/day 1-21). The CR/CRi rates in the two arms were not different (69 vs. 66%). Event-free survival (EFS) at 36 months was 19% for the standard arm versus 21% for the lenalidomide arm and overall survival (OS) 35% vs. 30%, respectively. The frequencies and grade of adverse events were not significantly different between the treatment arms. Cardiovascular toxicities were rare and equally distributed between the arms. The results of the present study show that the addition of lenalidomide to standard remission induction chemotherapy does not improve the therapeutic outcome of older AML patients. This trial is registered as number NTR2294 in The NederlandsTrial Register (www.trialregister.nl).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Induction Chemotherapy/mortality , Leukemia, Myeloid, Acute/drug therapy , Myelodysplastic Syndromes/drug therapy , Aged , Aged, 80 and over , Cytarabine/administration & dosage , Female , Follow-Up Studies , Humans , Lenalidomide/administration & dosage , Leukemia, Myeloid, Acute/pathology , Male , Myelodysplastic Syndromes/pathology , Prognosis , Remission Induction , Survival Rate
7.
Ther Adv Hematol ; 7(6): 330-344, 2016 Dec.
Article in English | MEDLINE | ID: mdl-27904737

ABSTRACT

Even though the prognosis of patients with multiple myeloma is continuing to improve, all patients eventually develop relapsed refractory disease. Several novel therapeutics have been developed in the last few years including the second-generation proteasome inhibitor carfilzomib which has been approved for patients with relapsed and refractory multiple myeloma in the United States since 2012. Recently data from several phase III studies have become available showing the promising efficacy of carfilzomib in combination with lenalidomide, which led to the renewed approval of carfilzomib in combination with lenalidomide and dexamethasone for relapsed myeloma in 2015. Furthermore carfilzomib showed superiority over bortezomib on both efficacy and toxicity profiles, especially a profoundly lower incidence in polyneuropathy. Carfilzomib has been shown to partially overcome the negative effects of high-risk cytogenetics. Promising combinations of carfilzomib with histone deacetylase (HDAC) inhibitors, pomalidomide and several other novel therapeutics have been presented in early studies. The optimal dosing regimen and sequence of treatment regimens remain important questions for the future.

8.
Leukemia ; 30(2): 439-46, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26437777

ABSTRACT

Relapses after initial successful treatment in acute myeloid leukemia are thought to originate from the outgrowth of leukemic stem cells. Their flow cytometrically assessed frequency is of importance for relapse prediction and is therefore assumed to be implemented in future risk group profiling. Since current detection methods are complex, time- and bone marrow consuming (multiple-tubes approach), it would be advantageous to have a broadly applicable approach that enables to quantify leukemia stem cells both at diagnosis and follow-up. We compared 15 markers in 131 patients concerning their prevalence, usefulness and stability in CD34(+)CD38(-) leukemic stem cell detection in healthy controls, acute myeloid leukemia diagnosis and follow-up samples. Ultimately, we designed a single 8-color detection tube including common markers CD45, CD34 and CD38, and specific markers CD45RA, CD123, CD33, CD44 and a marker cocktail (CLL-1/TIM-3/CD7/CD11b/CD22/CD56) in one fluorescence channel. Validation analyses in 31 patients showed that the single tube approach was as good as the multiple-tube approach. Our approach requires the least possible amounts of bone marrow, and is suitable for multi-institutional studies. Moreover, it enables detection of leukemic stem cells both at time of diagnosis and follow-up, thereby including initially low-frequency populations emerging under therapy pressure.


Subject(s)
Leukemia, Myeloid, Acute/pathology , Neoplastic Stem Cells/immunology , ADP-ribosyl Cyclase 1/analysis , Antigens, CD34/analysis , Humans , Immunophenotyping , Interleukin-3 Receptor alpha Subunit/analysis , Membrane Glycoproteins/analysis , Sialic Acid Binding Ig-like Lectin 3/analysis
10.
Leukemia ; 30(3): 708-15, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26373238

ABSTRACT

As relapses are common in acute myeloid leukemia (AML), early relapse prediction is of high importance. Although conventional minimal residual disease (MRD) measurement is carried out in bone marrow (BM), peripheral blood (PB) would be an advantageous alternative source. This study aims to investigate the specificity of leukemia-associated immunophenotypes used for MRD detection in blood samples. Consistency of PB MRD as compared with BM MRD was determined in flow cytometric data of 205 paired BM and PB samples of 114 AML patients. A significant correlation was found between PB and BM MRD (r=0.67, P<0.001), while median PB MRD percentage was factor 4-5 lower compared with BM MRD. Primitive blast (CD34+/CD117+/CD133+) frequency was significantly lower in PB (median factor 23.7), indicating that PB MRD detection is more specific than BM. Cumulative incidence of relapse 1 year after induction therapy was 29% for PB MRD-negative and 89% for PB MRD-positive patients (P<0.001). Three-year OS was 52% for MRD-negative and 15% for MRD-positive patients (P=0.034). Similar differences were found after consolidation therapy. As PB MRD appeared to be an independent predictor for response duration, the highly specific PB MRD assay may have a prominent role in future MRD assessment in AML.


Subject(s)
Bone Marrow/pathology , Leukemia, Myeloid, Acute/diagnosis , Leukocytes, Mononuclear/pathology , Adult , Aged , Antigens, CD/immunology , Antineoplastic Agents/therapeutic use , Biomarkers/analysis , Bone Marrow/immunology , Case-Control Studies , Consolidation Chemotherapy/methods , Female , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/mortality , Leukemia, Myeloid, Acute/pathology , Leukocytes, Mononuclear/immunology , Male , Middle Aged , Neoplasm, Residual , Prognosis , Recurrence , Survival Analysis
11.
Article in French | MEDLINE | ID: mdl-29468863

ABSTRACT

Benzodiazepine hypnotics bear a higher risk of high dose dependence than benzodiazepine anxiolytics, according to a recent study in Luxemburg. This article summarizes the main indications of these molecules and the current treatment recommendations. It provides an overview of public health actions of the past and the future to reduce their excessive consumption.


Subject(s)
Anxiety/drug therapy , Benzodiazepines/therapeutic use , Sleep Initiation and Maintenance Disorders/drug therapy , Humans , Practice Guidelines as Topic
12.
Mol Cancer Res ; 13(6): 982-92, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25944917

ABSTRACT

UNLABELLED: Loss of ephrin receptor (EphB1) expression may associate with aggressive cancer phenotypes; however, the mechanism of action remains unclear. To gain detailed insight into EphB1 function in acute myelogenous leukemia (AML), comprehensive analysis of EphB1 transcriptional regulation was conducted. In AML cells, EphB1 transcript was inversely correlated with EphB1 promoter methylation. The presence of EphB1 allowed EfnB1 ligand-mediated p53 DNA binding, leading to restoration of the DNA damage response (DDR) cascade by the activation of ATR, Chk1, p53, p21, p38, CDK1(tyr15), and Bax, and downregulation of HSP27 and Bcl2. Comparatively, reintroduction of EphB1 expression in EphB1-methylated AML cells enhanced the same cascade of ATR, Chk1, p21, and CDK1(tyr15), which consequently enforced programmed cell death. Interestingly, in pediatric AML samples, EphB1 peptide phosphorylation and mRNA expression were actively suppressed as compared with normal bone marrow, and a significant percentage of the primary AML specimens had EphB1 promoter hypermethylation. Finally, EphB1 repression associated with a poor overall survival in pediatric AML. Combined, the contribution of EphB1 to the DDR system reveals a tumor-suppressor function for EphB1 in pediatric AML. IMPLICATIONS: The tumor-suppressor function of EphB1 is clinically relevant across many malignancies, suggesting that EphB1 is an important regulator of common cancer cell transforming pathways.


Subject(s)
DNA Damage , Down-Regulation , Leukemia, Myeloid, Acute/metabolism , Receptor, EphB2/metabolism , Apoptosis , Bone Marrow , Cell Line, Tumor , Child , DNA Methylation , DNA Repair , G2 Phase Cell Cycle Checkpoints , Humans , Leukemia, Myeloid, Acute/pathology , Promoter Regions, Genetic , Receptor, EphA1/metabolism
13.
Rev Med Liege ; 68(5-6): 303-10, 2013.
Article in French | MEDLINE | ID: mdl-23888581

ABSTRACT

Benzodiazepines have been considered the treatment of choice for a variety of neuropsychiatric disorders. They are currently much more controversial and drugs considered less dangerous are generally preferred. This article summarizes the characteristics of the different benzodiazepines present on the Belgian market. It describes abuse and dependence, as well as the risks of these substances in specific populations or situations. New data suggest that there is a much higher risk of decease in case of a chronic use. Finally, recommendations on rational use and withdrawal are given.


Subject(s)
Benzodiazepines/administration & dosage , Benzodiazepines/adverse effects , Anxiety Disorders/drug therapy , Ataxia/chemically induced , Cognition Disorders/chemically induced , Depression/drug therapy , Fatigue/chemically induced , Humans , Neoplasms/chemically induced , Sleep Wake Disorders/drug therapy , Substance-Related Disorders/complications , Substance-Related Disorders/etiology
14.
Neuropathol Appl Neurobiol ; 39(4): 426-36, 2013 Jun.
Article in English | MEDLINE | ID: mdl-22845849

ABSTRACT

AIMS: Diffuse intrinsic pontine glioma (DIPG) is a fatal paediatric malignancy. Tumour resection is not possible without serious morbidity and biopsies are rarely performed. The resulting lack of primary DIPG material has made preclinical research practically impossible and has hindered the development of new therapies for this disease. The aim of the current study was to address the lack of primary DIPG material and preclinical models by developing a multi-institutional autopsy protocol. METHODS: An autopsy protocol was implemented in the Netherlands to obtain tumour material within a brief post mortem interval. A team of neuropathologists and researchers was available at any time to perform the autopsy and process the material harvested. Whole brain autopsy was performed and primary DIPG material and healthy tissue were collected from all affected brain areas. Finally, the study included systematic evaluation by parents. RESULTS: Five autopsies were performed. The mean time interval between death and time of autopsy was 3 h (range 2-4). All tumours were graded as glioblastoma. None of the parents regretted their choice to participate, and they all derived comfort in donating tissue of their child in the hope to help future DIPG patients. In addition, we developed and characterized one of the first DIPG cell cultures from post mortem material. CONCLUSION: Here we show that obtaining post mortem DIPG tumour tissue for research purposes is feasible with short delay, and that the autopsy procedure is satisfying for participating parents and can be suitable for the development of preclinical DIPG models.


Subject(s)
Autopsy/standards , Brain Stem Neoplasms/pathology , Glioma/pathology , Primary Cell Culture/standards , Animals , Child , Child, Preschool , DNA, Neoplasm/biosynthesis , DNA, Neoplasm/genetics , Female , Humans , Infant , Male , Mice , Mice, Nude , Parents , Pons/pathology , RNA, Neoplasm/biosynthesis , RNA, Neoplasm/genetics
15.
Leukemia ; 26(6): 1313-20, 2012 Jun.
Article in English | MEDLINE | ID: mdl-22289983

ABSTRACT

The majority of pediatric and younger adult (<60 years) AML patients achieve complete remission. However, 30-40% of patients relapse and display a dismal outcome. Recently we described a frequent instability of type I/II mutations between diagnosis and relapse. Here, we explored the hypothesis that these mutational shifts originate from clonal selection during treatment/disease progression. Subfractions of blasts from initial diagnosis samples were cell sorted and their mutational profiles were compared with those of the corresponding relapse samples of 7 CD34(+) AML patients. At diagnosis, subfractions of the CD45(dim)CD34(+)CD38(dim/-) compartment were heterogeneous in the distribution of mutations, when compared to the whole CD45(dim)CD34(+) blast compartment in 6 out of 7 patients. Moreover, within CD45(dim)CD34(+)CD38(dim/-) fraction of initial samples of 5 of these 6 AML patients, we found evidence for the presence of a minor, initially undetected subpopulation with a specific mutational profile that dominated the bulk of leukemic blasts at relapse. In conclusion, our findings lend support to the AML oligoclonality concept and provide molecular evidence for selection and expansion of a chemo-resistant subpopulation towards development of relapse. These results imply that early detection of pre-existing drug-resistant leukemic subpopulations is crucial for relapse prevention by proper timing of targeted treatment.


Subject(s)
ADP-ribosyl Cyclase 1/metabolism , Antigens, CD34/metabolism , Biomarkers, Tumor/genetics , Leukemia, Myeloid, Acute/diagnosis , Neoplasm Recurrence, Local/diagnosis , Neoplasm Recurrence, Local/genetics , Adolescent , Adult , Clone Cells , DNA Mutational Analysis , Female , Flow Cytometry , Genes, ras/genetics , Humans , Immunophenotyping , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Male , Middle Aged , Mutation/genetics , Neoplasm Recurrence, Local/metabolism , Nuclear Proteins/genetics , Nucleophosmin , Prognosis , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins p21(ras) , Remission Induction , WT1 Proteins/genetics , fms-Like Tyrosine Kinase 3/genetics , ras Proteins/genetics
16.
Leukemia ; 26(4): 757-68, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21941364

ABSTRACT

Proteasome inhibition is a novel treatment for several hematological malignancies. However, resistance to the proteasome inhibitor bortezomib (BTZ, Velcade) is an emerging clinical impediment. Mutations in the ß5 subunit of the proteasome, the primary target of BTZ, have been associated with drug resistance. However, the exact mechanism by which these mutations contribute to BTZ resistance, is still largely unknown. Toward this end, we here developed BTZ-resistant multiple myeloma (8226) and acute lymphoblastic leukemia (CCRF-CEM) cell line models by exposure to stepwise increasing concentrations of BTZ. Characterization of the various BTZ-resistant cells revealed upregulation of mutant ß5 subunit of the proteasome. These newly identified ß5-subunit mutations, along with previously described mutations, formed a mutation cluster region in the BTZ-binding pocket of the ß5 subunit, that of the S1 specificity pocket in particular. Moreover, we provide the first evidence that the mechanism underlying BTZ resistance in these tumor cells is impaired binding of BTZ to the mutant ß5 subunit of the proteasome. We propose that proteasome subunit overexpression is an essential compensatory mechanism for the impaired catalytic activity of these mutant proteasomes. Our findings further suggest that second-generation proteasome inhibitors that target the α7 subunit of the proteasome can overcome this drug resistance modality.


Subject(s)
Antineoplastic Agents/metabolism , Boronic Acids/metabolism , Multiple Myeloma/drug therapy , Mutation , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Proteasome Endopeptidase Complex/genetics , Pyrazines/metabolism , Amino Acid Substitution , Boronic Acids/therapeutic use , Bortezomib , Cell Line, Tumor , Drug Resistance, Neoplasm , Humans , Multiple Myeloma/genetics , Multiple Myeloma/pathology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/genetics , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Proteasome Endopeptidase Complex/metabolism , Proteasome Inhibitors , Pyrazines/therapeutic use
17.
Leukemia ; 23(2): 262-70, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19020547

ABSTRACT

Nucleophosmin (NPM1) mutations occur frequently in adult cytogenetically normal acute myeloid leukemia (CN-AML) and confer favorable outcome. We investigated the frequency and prognostic significance of NPM1 mutations in childhood AML (n=298), specifically focusing on the CN-AML subgroup (n=100). Mutations were found in 8.4%, and clustered significantly in the CN-AML subgroup (22%). No mutations were found in patients below the age of 3 years; in CN-AML, there was an increasing incidence above this age. In the overall group, NPM1 mutations conferred an independent favorable prognostic impact on event-free survival (5-year pEFS 66 vs 39%; P=0.02), which did not translate into a significantly better overall survival (5-year pOS 68 vs 56%; P=0.30). However, when the favorable cytogenetic subgroups [inv(16) and t(8;21)] were excluded from the NPM1 wild-type group, the difference in pOS was borderline statistically significant (68 vs 45%; P=0.07). In the CN-AML cohort, NPM1 mutations were an independent prognostic factor on pEFS (80 vs 39%; P=0.01), and pOS (85 vs 60%; P=0.06), which was not influenced by FLT3/ITD. However, in NPM1 wild-type CN-AML, FLT3/ITD-positive patients had a significantly worse outcome (pEFS 48 vs 18%; P<0.001). We conclude that NPM1 mutations confer a favorable prognosis in childhood AML and in CN-AML in particular.


Subject(s)
Leukemia, Myeloid, Acute/genetics , Mutation , Nuclear Proteins/genetics , Adolescent , Age Factors , Child , Child, Preschool , Cytogenetic Analysis , Female , Humans , Infant , Leukemia, Myeloid, Acute/diagnosis , Male , Nucleophosmin , Predictive Value of Tests , Prognosis , Retrospective Studies , Survival Analysis
18.
Cell Oncol ; 30(4): 299-306, 2008.
Article in English | MEDLINE | ID: mdl-18607065

ABSTRACT

OBJECTIVE: Inactivation of the FA-BRCA pathway results in chromosomal instability. Fanconi anaemia (FA) patients have an inherited defect in this pathway and are strongly predisposed to the development of acute myeloid leukaemia (AML). Studies in sporadic cancers have shown promoter methylation of the FANCF gene in a significant proportion of various solid tumours. However, only a single leukaemic case with methylation of one of the FA-BRCA genes has been described to date, i.e. methylation of FANCF in cell line CHRF-288. We investigated the presence of aberrant methylation in 11 FA-BRCA genes in sporadic cases of leukaemia. METHODS: We analyzed promoter methylation in 143 AML bone marrow samples and 97 acute lymphoblastic leukaemia (ALL) samples using methylation-specific multiplex ligation-dependent probe amplification (MS-MLPA). Samples with aberrant methylation were further analyzed by bisulphite sequencing and tested for mitomycin C sensitivity using Colony Forming Units assays. RESULTS: MS-MLPA showed promoter methylation of FANCC in one AML and three ALL samples, while FANCL was found methylated in one ALL sample. Bisulphite sequencing of promoter regions confirmed hypermethylation in all cases. In addition, samples with hypermethylation of either FANCC or FANCL appeared more sensitive towards mitomycin C in Colony Forming Units assays, compared to controls. CONCLUSION: Hypermethylation of promoter regions from FA-BRCA genes does occur in sporadic leukaemia, albeit infrequently. Hypermethylation was found to result in hypersensitivity towards DNA cross-linking agents, a hallmark of the FA cellular phenotype, suggesting that these samples displayed chromosomal instability. This instability may have contributed to the occurrence of the leukaemia. In addition, this is the first report to describe hypermethylation of FANCC and FANCL. This warrants the investigation of multiple FA-BRCA genes in other malignancies.


Subject(s)
Bone Marrow Cells/enzymology , DNA Methylation , Fanconi Anemia Complementation Group C Protein/genetics , Fanconi Anemia Complementation Group L Protein/genetics , Leukemia, Myeloid, Acute/genetics , Precursor Cell Lymphoblastic Leukemia-Lymphoma/genetics , Promoter Regions, Genetic/genetics , Adult , Bone Marrow Cells/pathology , Child , Child, Preschool , Cross-Linking Reagents/therapeutic use , DNA Modification Methylases/genetics , DNA Modification Methylases/metabolism , Fanconi Anemia Complementation Group Proteins/genetics , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/pathology , Mitomycin/therapeutic use , Precursor Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Precursor Cell Lymphoblastic Leukemia-Lymphoma/pathology , Tumor Stem Cell Assay
20.
Br J Haematol ; 134(3): 253-62, 2006 Aug.
Article in English | MEDLINE | ID: mdl-16787504

ABSTRACT

Following its success in multiple myeloma (MM), proteasome inhibition has become a topic of interest as novel treatment strategy of cancer. By simultaneously affecting multiple pathways in the cancer cell, such as deregulation of the programmed degradation of many cellular proteins, proteasome inhibition causes rapid apoptosis of these cells. Both in rapidly proliferating leukaemic cell lines and in primary leukaemic cells isolated from patients, proteasome inhibition results in antileukaemic activity. The normal counterparts of these cells are much more resistant to proteasome inhibitors (PI), thereby resulting in a favourable therapeutic index. Importantly, while leukaemic stem cells are sensitive to proteasome inhibition, normal haematopoietic stem cells are still viable after drug exposure. Nowadays, many PIs are being identified; bortezomib is the most well known since obtaining Food and Drug Administration approval for clinical use in MM. This review summarises the biological and clinical aspects of proteasome inhibition and discusses the potential role of these inhibitors in the treatment of leukaemia.


Subject(s)
Antineoplastic Agents/therapeutic use , Boronic Acids/therapeutic use , Leukemia/drug therapy , Protease Inhibitors/therapeutic use , Pyrazines/therapeutic use , Apoptosis/drug effects , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Clinical Trials, Phase I as Topic , Forecasting , Humans , Leukemia/metabolism , NF-kappa B/metabolism , Ubiquitin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL