Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 46
Filtrer
1.
J Clin Oncol ; 42(10): 1158-1168, 2024 Apr 01.
Article de Anglais | MEDLINE | ID: mdl-38215358

RÉSUMÉ

PURPOSE: To determine the optimal induction chemotherapy regimen for younger adults with newly diagnosed AML without known adverse risk cytogenetics. PATIENTS AND METHODS: One thousand thirty-three patients were randomly assigned to intensified (fludarabine, cytarabine, granulocyte colony-stimulating factor, and idarubicin [FLAG-Ida]) or standard (daunorubicin and Ara-C [DA]) induction chemotherapy, with one or two doses of gemtuzumab ozogamicin (GO). The primary end point was overall survival (OS). RESULTS: There was no difference in remission rate after two courses between FLAG-Ida + GO and DA + GO (complete remission [CR] + CR with incomplete hematologic recovery 93% v 91%) or in day 60 mortality (4.3% v 4.6%). There was no difference in OS (66% v 63%; P = .41); however, the risk of relapse was lower with FLAG-Ida + GO (24% v 41%; P < .001) and 3-year event-free survival was higher (57% v 45%; P < .001). In patients with an NPM1 mutation (30%), 3-year OS was significantly higher with FLAG-Ida + GO (82% v 64%; P = .005). NPM1 measurable residual disease (MRD) clearance was also greater, with 88% versus 77% becoming MRD-negative in peripheral blood after cycle 2 (P = .02). Three-year OS was also higher in patients with a FLT3 mutation (64% v 54%; P = .047). Fewer transplants were performed in patients receiving FLAG-Ida + GO (238 v 278; P = .02). There was no difference in outcome according to the number of GO doses, although NPM1 MRD clearance was higher with two doses in the DA arm. Patients with core binding factor AML treated with DA and one dose of GO had a 3-year OS of 96% with no survival benefit from FLAG-Ida + GO. CONCLUSION: Overall, FLAG-Ida + GO significantly reduced relapse without improving OS. However, exploratory analyses show that patients with NPM1 and FLT3 mutations had substantial improvements in OS. By contrast, in patients with core binding factor AML, outcomes were excellent with DA + GO with no FLAG-Ida benefit.


Sujet(s)
Idarubicine , Leucémie aigüe myéloïde , Vidarabine/analogues et dérivés , Tyrosine kinase-3 de type fms , Adulte , Humains , Gemtuzumab/usage thérapeutique , Facteur de stimulation des colonies de granulocytes/usage thérapeutique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/diagnostic , Survie sans progression , Cytarabine/usage thérapeutique , Récidive tumorale locale/traitement médicamenteux , Vidarabine/usage thérapeutique , Protéines nucléaires/génétique , Mutation , Facteurs de transcription CBF , Récidive , Protocoles de polychimiothérapie antinéoplasique/effets indésirables
2.
Blood Adv ; 8(2): 343-352, 2024 01 23.
Article de Anglais | MEDLINE | ID: mdl-38039513

RÉSUMÉ

ABSTRACT: Molecular failure in NPM1-mutated acute myeloid leukemia (AML) inevitably progresses to frank relapse if untreated. Recently published small case series show that venetoclax combined with low-dose cytarabine or azacitidine can reduce or eliminate measurable residual disease (MRD). Here, we report on an international multicenter cohort of 79 patients treated for molecular failure with venetoclax combinations and report an overall molecular response (≥1-log reduction in MRD) in 66 patients (84%) and MRD negativity in 56 (71%). Eighteen of 79 patients (23%) required hospitalization, and no deaths were reported during treatment. Forty-one patients were bridged to allogeneic transplant with no further therapy, and 25 of 41 were MRD negative assessed by reverse transcription quantitative polymerase chain reaction before transplant. Overall survival (OS) for the whole cohort at 2 years was 67%, event-free survival (EFS) was 45%, and in responding patients, there was no difference in survival in those who received a transplant using time-dependent analysis. Presence of FLT3-ITD mutation was associated with a lower response rate (64 vs 91%; P < .01), worse OS (hazard ratio [HR], 2.50; 95% confidence interval [CI], 1.06-5.86; P = .036), and EFS (HR, 1.87; 95% CI, 1.06-3.28; P = .03). Eighteen of 35 patients who did not undergo transplant became MRD negative and stopped treatment after a median of 10 months, with 2-year molecular relapse free survival of 62% from the end of treatment. Venetoclax-based low intensive chemotherapy is a potentially effective treatment for molecular relapse in NPM1-mutated AML, either as a bridge to transplant or as definitive therapy.


Sujet(s)
Composés hétérocycliques bicycliques , Leucémie aigüe myéloïde , Protéines nucléaires , Sulfonamides , Humains , Composés hétérocycliques bicycliques/pharmacologie , Composés hétérocycliques bicycliques/usage thérapeutique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Mutation , Protéines nucléaires/génétique , Nucléophosmine/génétique , Récidive , Sulfonamides/pharmacologie , Sulfonamides/usage thérapeutique
3.
Nat Commun ; 14(1): 7725, 2023 Nov 25.
Article de Anglais | MEDLINE | ID: mdl-38001082

RÉSUMÉ

Current therapies for myeloproliferative neoplasms (MPNs) improve symptoms but have limited effect on tumor size. In preclinical studies, tamoxifen restored normal apoptosis in mutated hematopoietic stem/progenitor cells (HSPCs). TAMARIN Phase-II, multicenter, single-arm clinical trial assessed tamoxifen's safety and activity in patients with stable MPNs, no prior thrombotic events and mutated JAK2V617F, CALRins5 or CALRdel52 peripheral blood allele burden ≥20% (EudraCT 2015-005497-38). 38 patients were recruited over 112w and 32 completed 24w-treatment. The study's A'herns success criteria were met as the primary outcome ( ≥ 50% reduction in mutant allele burden at 24w) was observed in 3/38 patients. Secondary outcomes included ≥25% reduction at 24w (5/38), ≥50% reduction at 12w (0/38), thrombotic events (2/38), toxicities, hematological response, proportion of patients in each IWG-MRT response category and ELN response criteria. As exploratory outcomes, baseline analysis of HSPC transcriptome segregates responders and non-responders, suggesting a predictive signature. In responder HSPCs, longitudinal analysis shows high baseline expression of JAK-STAT signaling and oxidative phosphorylation genes, which are downregulated by tamoxifen. We further demonstrate in preclinical studies that in JAK2V617F+ cells, 4-hydroxytamoxifen inhibits mitochondrial complex-I, activates integrated stress response and decreases pathogenic JAK2-signaling. These results warrant further investigation of tamoxifen in MPN, with careful consideration of thrombotic risk.


Sujet(s)
Syndromes myéloprolifératifs , Tumeurs , Humains , Syndromes myéloprolifératifs/traitement médicamenteux , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/anatomopathologie , Kinase Janus-2/génétique , Kinase Janus-2/métabolisme , Cellules souches hématopoïétiques/métabolisme , Transduction du signal , Tumeurs/métabolisme , Tamoxifène/usage thérapeutique , Tamoxifène/métabolisme , Mutation , Calréticuline/génétique , Calréticuline/métabolisme
4.
Blood ; 142(20): 1697-1707, 2023 11 16.
Article de Anglais | MEDLINE | ID: mdl-37595359

RÉSUMÉ

Addition of gemtuzumab ozogamicin (GO) to induction chemotherapy improves outcomes in older patients with acute myeloid leukemia (AML), but it is uncertain whether a fractionated schedule provides additional benefit to a single dose. We randomized 852 older adults (median age, 68-years) with AML/high-risk myelodysplasia to GO on day 1 (GO1) or on days 1 and 4 (GO2) of course 1 induction. The median follow-up period was 50.2 months. Although complete remission (CR) rates after course 1 did not significantly differ between arms (GO2, 63%; GO1, 57%; odds ratio [OR], 0.78; P = .08), there were significantly more patients who achieved CR with a measurable residual disease (MRD)<0.1% (50% vs 41%; OR, 0.72; P = .027). This differential MRD reduction with GO2 varied across molecular subtypes, being greatest for IDH mutations. The 5-year overall survival (OS) was 29% for patients in the GO2 arm and 24% for those in the GO1 arm (hazard ratio [HR], 0.89; P = .14). In a sensitivity analysis excluding patients found to have adverse cytogenetics or TP53 mutations, the 5-year OS was 33% for GO2 and 26% for GO1 (HR, 0.83; P = .045). In total, 228 (27%) patients received an allogeneic transplantation in first remission. Posttransplant OS was superior in the GO2 arm (HR, 0.67; P = .033); furthermore, the survival advantage from GO2 in the sensitivity analysis was lost when data of patients were censored at transplantation. In conclusion, GO2 was associated with a greater reduction in MRD and improved survival in older adults with nonadverse risk genetics. This benefit from GO2 was dependent on allogeneic transplantation to translate the better leukemia clearance into improved survival. This trial was registered at www.isrctn.com as #ISRCTN 31682779.


Sujet(s)
Daunorubicine , Leucémie aigüe myéloïde , Humains , Sujet âgé , Gemtuzumab/usage thérapeutique , Anticorps monoclonaux humanisés , Cytarabine , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Royaume-Uni , Aminosides , Protocoles de polychimiothérapie antinéoplasique/effets indésirables
5.
Leukemia ; 37(10): 2066-2072, 2023 10.
Article de Anglais | MEDLINE | ID: mdl-37558736

RÉSUMÉ

Patients with FLT3-mutated AML have a high relapse rate and suboptimal outcomes. Many have co-mutations suitable for measurable residual disease (MRD) monitoring by RT-qPCR and those destined to relapse can be identified by high or rising levels of MRD, called molecular failure.  This provides a window for pre-emptive intervention, but there is little evidence to guide treatment. The use of FLT3 inhibitors (FLT3i) appears attractive but their use has not yet been evaluated.  We identified 56 patients treated with FLT3i at molecular failure.  The FLT3 mutation was an ITD in 52, TKD in 7 and both in 3. Over half of patients had previously received midostaurin. Molecular failure occurred at a median 9.2 months from diagnosis and was treated with gilteritinib (n = 38), quizartinib (n = 7) or sorafenib (n = 11). 60% achieved a molecular response, with 45% reaching MRD negativity. Haematological toxicity was low, and 22 patients were bridged directly to allogeneic transplant with another 6 to donor lymphocyte infusion. 2-year overall survival was 80% (95%CI 69-93) and molecular event-free survival 56% (95%CI 44-72). High-sensitivity next-generation sequencing for FLT3-ITD at molecular failure identified patients more likely to benefit. FLT3i monotherapy for molecular failure is a promising strategy which merits evaluation in prospective studies.


Sujet(s)
Leucémie aigüe myéloïde , Thérapie de rattrapage , Humains , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Tyrosine kinase-3 de type fms/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Mutation , Récidive tumorale locale , Études prospectives , Inhibiteurs de protéines kinases/usage thérapeutique
6.
J Clin Oncol ; 41(19): 3534-3544, 2023 07 01.
Article de Anglais | MEDLINE | ID: mdl-37126762

RÉSUMÉ

PURPOSE: Polycythemia vera (PV) is characterized by JAK/STAT activation, thrombotic/hemorrhagic events, systemic symptoms, and disease transformation. In high-risk PV, ruxolitinib controls blood counts and improves symptoms. PATIENTS AND METHODS: MAJIC-PV is a randomized phase II trial of ruxolitinib versus best available therapy (BAT) in patients resistant/intolerant to hydroxycarbamide (HC-INT/RES). Primary outcome was complete response (CR) within 1 year. Secondary outcomes included duration of response, event-free survival (EFS), symptom, and molecular response. RESULTS: One hundred eighty patients were randomly assigned. CR was achieved in 40 (43%) patients on ruxolitinib versus 23 (26%) on BAT (odds ratio, 2.12; 90% CI, 1.25 to 3.60; P = .02). Duration of CR was superior for ruxolitinib (hazard ratio [HR], 0.38; 95% CI, 0.24 to 0.61; P < .001). Symptom responses were better with ruxolitinib and durable. EFS (major thrombosis, hemorrhage, transformation, and death) was superior for patients attaining CR within 1 year (HR, 0.41; 95% CI, 0.21 to 0.78; P = .01); and those on ruxolitinib (HR, 0.58; 95% CI, 0.35 to 0.94; P = .03). Serial analysis of JAK2V617F variant allele fraction revealed molecular response was more frequent with ruxolitinib and was associated with improved outcomes (progression-free survival [PFS] P = .001, EFS P = .001, overall survival P = .01) and clearance of JAK2V617F stem/progenitor cells. ASXL1 mutations predicted for adverse EFS (HR, 3.02; 95% CI, 1.47 to 6.17; P = .003). The safety profile of ruxolitinib was as previously reported. CONCLUSION: The MAJIC-PV study demonstrates ruxolitinib treatment benefits HC-INT/RES PV patients with superior CR, and EFS as well as molecular response; importantly also demonstrating for the first time, to our knowledge, that molecular response is linked to EFS, PFS, and OS.


Sujet(s)
Polyglobulie primitive essentielle , Humains , Polyglobulie primitive essentielle/traitement médicamenteux , Polyglobulie primitive essentielle/génétique , Polyglobulie primitive essentielle/complications , Résultat thérapeutique , Hydroxy-urée/effets indésirables , Nitriles/usage thérapeutique , Hémorragie/complications , Hémorragie/traitement médicamenteux
7.
Br J Haematol ; 200(5): 573-578, 2023 03.
Article de Anglais | MEDLINE | ID: mdl-36413792

RÉSUMÉ

The survival of acute myeloid leukaemia (AML) patients aged over 60 has been suboptimal historically, whether they are treated using hypomethylating agents, low-dose cytarabine (LDAC) or venetoclax-based regimens. Progress is being made, however, for subgroups with favourable molecular or cytogenetic findings. Arginine metabolism plays a key role in AML pathophysiology. We report the only randomised study of LDAC with recombinant arginase BCT-100 versus LDAC alone in older AML patients unsuitable for intensive therapy. Eighty-three patients were randomised to the study. An overall response rate was seen in 19.5% (all complete remission [CR]) and 15% (7.5% each in CR and CR without evidence of adequate count recovery [CRi]) of patients in the LDAC+BCT-100 and LDAC arms respectively (odds ratio 0.73, confidence interval 0.23-2.33; p = 0.592). No significant difference in overall or median survival between treatment arms was seen. The addition of BCT-100 to LDAC was well tolerated.


Sujet(s)
Cytarabine , Leucémie aigüe myéloïde , Humains , Adulte d'âge moyen , Sujet âgé , Arginase , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Polyéthylène glycols/usage thérapeutique
10.
Blood Adv ; 5(24): 5621-5625, 2021 12 28.
Article de Anglais | MEDLINE | ID: mdl-34597366

RÉSUMÉ

Survival for older patients with acute myeloid leukemia (AML) unsuitable for intensive chemotherapy is unsatisfactory. Standard nonintensive therapies have low response rates and only extend life by a few months. Quizartinib is an oral Fms-like tyrosine kinase 3 (FLT3) inhibitor with reported activity in wild-type patients. As part of the AML LI trial, we undertook a randomized evaluation of low-dose ara-C (LDAC) with or without quizartinib in patients not fit for intensive chemotherapy. Overall, survival was not improved (202 patients), but in the 27 FLT3-ITD patients, the addition of quizartinib to LDAC improved response (P = .05) with complete remission/complete remission with incomplete haematological recovery for quizartinib + LDAC in 5/13 (38%) vs 0/14 (0%) in patients receiving LDAC alone. Overall survival (OS) in these FLT3-ITD+ patients was also significantly improved at 2 years for quizartinib + LDAC (hazard ratio 0.36; 95% confidence intervals: 0.16, 0.85, P = .04). Median OS was 13.7 months compared with 4.2 months with LDAC alone. This is the first report of an FLT3-targeted therapy added to standard nonintensive chemotherapy that has improved survival in this population. Quizartinib merits consideration for future triplet-based treatment approaches. This trial was registered at www.clinicaltrials.gov as ISRCTN #ISRCTN40571019 and EUDRACT @2011-000749-19.


Sujet(s)
Cytarabine , Leucémie aigüe myéloïde , Sujet âgé , Benzothiazoles , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Phénylurées
12.
Leukemia ; 35(1): 62-74, 2021 01.
Article de Anglais | MEDLINE | ID: mdl-32203138

RÉSUMÉ

Talacotuzumab, a humanized anti-CD123 monoclonal antibody, was evaluated in combination with decitabine in elderly patients with acute myeloid leukemia (AML) not eligible for intensive chemotherapy. A multicenter, phase 2/3 study was initiated to determine the recommended phase 2 dose (RP2D) of talacotuzumab (Part A) followed by an open-label, randomized comparison of talacotuzumab in combination with decitabine versus decitabine alone to assess achievement of complete response (CR) and overall survival (OS) in Part B. Ten patients were enrolled in Part A and 316 in Part B; the results presented here are based on a database lock on January 25, 2018. Part A confirmed the RP2D of talacotuzumab to be 9 mg/kg. In Part B, CR was achieved in 12/80 (15%) patients receiving combination therapy and in 9/82 (11%) patients receiving decitabine alone (odds ratio: 1.4; 95% confidence interval [CI]: 0.6-3.6; p = 0.44). Median (95% CI) OS was 5.36 (4.27-7.95) months for combination therapy versus 7.26 (6.47-8.64) months for decitabine alone (hazard ratio: 1.04; 95% CI: 0.79-1.37; p = 0.78). Combination therapy showed no improvement in efficacy versus decitabine alone, resulting in the Independent Data Monitoring Committee's recommendation of early termination of enrollment and discontinuation of talacotuzumab treatment.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Décitabine/usage thérapeutique , Sujet âgé , Anticorps monoclonaux humanisés/administration et posologie , Anticorps monoclonaux humanisés/pharmacocinétique , Antinéoplasiques immunologiques/administration et posologie , Antinéoplasiques immunologiques/pharmacocinétique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Décitabine/administration et posologie , Décitabine/effets indésirables , Décitabine/pharmacocinétique , Surveillance des médicaments , Femelle , Humains , Estimation de Kaplan-Meier , Leucémie aigüe myéloïde/traitement médicamenteux , Mâle , Adulte d'âge moyen , Pronostic , Plan de recherche , Résultat thérapeutique
13.
Leuk Lymphoma ; 62(2): 264-270, 2021 02.
Article de Anglais | MEDLINE | ID: mdl-33078658

RÉSUMÉ

In contrast to Fms-like tyrosine kinase 3 (FLT3), the influence of FLT3 ligand (FLT3L) on acute myeloid leukemia (AML) biology and disease prognosis has been poorly described. Here we provide an overview of the role played by FLT3L in AML. While being a cytokine implicated in the regulation of hematopoiesis, both in normal situation and after intensive chemotherapy, FLT3L has also a role in enhancing proliferation, inhibiting apoptosis and conferring resistance to FLT3 inhibitors in AML. Moreover, recent independent data show how its measurement may be helpful in the disease management. Indeed, FLT3L could provide a low cost, rapid and noninvasive assessment of chemosensitivity and blast clearance that has robust prognostic significance for patients with AML.


Sujet(s)
Leucémie aigüe myéloïde , Protéines membranaires , Hématopoïèse , Humains , Leucémie aigüe myéloïde/diagnostic , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Protéines membranaires/génétique , Mutation , Pronostic , Tyrosine kinase-3 de type fms/génétique
14.
Br J Haematol ; 193(1): 155-159, 2021 04.
Article de Anglais | MEDLINE | ID: mdl-32996123

RÉSUMÉ

Treatment of relapsed/resistant acute myeloid leukaemia (AML) remains a significant area of unmet patient need, the outlook for most patients remaining extremely poor. A promising approach is to augment the anti-tumour immune response in these patients; most cancers do not activate immune effector cells because they express immunosuppressive ligands. We have previously shown that CD200 (an immunosuppressive ligand) is overexpressed in AML and confers an inferior overall survival compared to CD200low/neg patients. Here we show that a fully human anti-CD200 antibody (TTI-CD200) can block the interaction of CD200 with its receptor and restore AML immune responses in vitro and in vivo.


Sujet(s)
Anticorps bloquants/immunologie , Antigènes CD/immunologie , Antinéoplasiques immunologiques/usage thérapeutique , Immunité/immunologie , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/thérapie , Animaux , Anticorps bloquants/pharmacologie , Antigènes CD/effets des médicaments et des substances chimiques , Études cas-témoins , Cellules CIK/immunologie , Humains , Immunité/effets des médicaments et des substances chimiques , Immunosuppression thérapeutique/méthodes , Leucémie aigüe myéloïde/mortalité , Ligands , Souris , Modèles animaux , Prévention secondaire/méthodes , Transplantation hétérologue/méthodes
15.
Hum Mol Genet ; 29(20): 3341-3349, 2020 12 18.
Article de Anglais | MEDLINE | ID: mdl-32959868

RÉSUMÉ

Many medical treatments, from oncology to psychiatry, can lower white blood cell counts and thus access to these treatments can be restricted to individuals with normal levels of white blood cells, principally in order to minimize risk of serious infection. This adversely affects individuals of African or Middle Eastern ancestries who have on average a reduced number of circulating white blood cells, because of the Duffy-null (CC) genotype at rs2814778 in the ACKR1 gene. Here, we investigate whether the Duffy-null genotype is associated with the risk of infection using the UK Biobank sample and the iPSYCH Danish case-cohort study, two population-based samples from different countries and age ranges. We found that a high proportion of those with the Duffy-null genotype (21%) had a neutrophil count below the threshold often used as a cut-off for access to relevant treatments, compared with 1% of those with the TC/TT genotype. In addition we found that despite its strong association with lower average neutrophil counts, the Duffy-null genotype was not associated with an increased risk of infection, viral or bacterial. These results have widespread implications for the clinical treatment of individuals of African ancestry and indicate that neutrophil thresholds to access treatments could be lowered in individuals with the Duffy-null genotype without an increased risk of infection.


Sujet(s)
/génétique , Système Duffy/génétique , Infections/étiologie , Polymorphisme de nucléotide simple , /génétique , Biobanques , Études de cohortes , Femelle , Génotype , Humains , Infections/anatomopathologie , Mâle , Adulte d'âge moyen
16.
Curr Cancer Drug Targets ; 20(7): 513-531, 2020.
Article de Anglais | MEDLINE | ID: mdl-32418523

RÉSUMÉ

Activating mutations of FMS-like tyrosine kinase 3 (FLT3) are present in 30% of acute myeloid leukaemia (AML) patients at diagnosis and confer an adverse clinical prognosis. Mutated FLT3 has emerged as a viable therapeutic target and a number of FLT3-directed tyrosine kinase inhibitors have progressed through clinical development over the last 10-15 years. The last two years have seen United States Food and Drug Administration (US FDA) approvals of the multi-kinase inhibitor midostaurin for newly-diagnosed FLT3-mutated patients, when used in combination with intensive chemotherapy, and of the more FLT3-selective agent gilteritinib, used as monotherapy, for patients with relapsed or treatment-refractory FLT3-mutated AML. The 'second generation' agents, quizartinib and crenolanib, are also at advanced stages of clinical development. Significant challenges remain in negotiating a variety of potential acquired drug resistance mechanisms and in optimizing sequencing of FLT3 inhibitory drugs with existing and novel treatment approaches in different clinical settings, including frontline therapy, relapsed/refractory disease, and maintenance treatment. In this review, the biology of FLT3, the clinical challenge posed by FLT3-mutated AML, the developmental history of the key FLT3-inhibitory compounds, mechanisms of disease resistance, and the future outlook for this group of agents, including current and planned clinical trials, is discussed.


Sujet(s)
Antinéoplasiques/administration et posologie , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Mutation , Inhibiteurs de protéines kinases/administration et posologie , Tyrosine kinase-3 de type fms/antagonistes et inhibiteurs , Tyrosine kinase-3 de type fms/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Hématopoïèse/physiologie , Humains , Leucémie aigüe myéloïde/métabolisme , Récidive , Résultat thérapeutique , Tyrosine kinase-3 de type fms/métabolisme
17.
Blood ; 135(9): 680-688, 2020 02 27.
Article de Anglais | MEDLINE | ID: mdl-31932839

RÉSUMÉ

Relapse remains the most common cause of treatment failure for patients with acute myeloid leukemia (AML) who undergo allogeneic stem cell transplantation (alloSCT), and carries a grave prognosis. Multiple studies have identified the presence of measurable residual disease (MRD) assessed by flow cytometry before alloSCT as a strong predictor of relapse, but it is not clear how these findings apply to patients who test positive in molecular MRD assays, which have far greater sensitivity. We analyzed pretransplant blood and bone marrow samples by reverse-transcription polymerase chain reaction in 107 patients with NPM1-mutant AML enrolled in the UK National Cancer Research Institute AML17 study. After a median follow-up of 4.9 years, patients with negative, low (<200 copies per 105ABL in the peripheral blood and <1000 copies in the bone marrow aspirate), and high levels of MRD had an estimated 2-year overall survival (2y-OS) of 83%, 63%, and 13%, respectively (P < .0001). Focusing on patients with low-level MRD before alloSCT, those with FLT3 internal tandem duplications(ITDs) had significantly poorer outcome (hazard ratio [HR], 6.14; P = .01). Combining these variables was highly prognostic, dividing patients into 2 groups with 2y-OS of 17% and 82% (HR, 13.2; P < .0001). T-depletion was associated with significantly reduced survival both in the entire cohort (2y-OS, 56% vs 96%; HR, 3.24; P = .0005) and in MRD-positive patients (2y-OS, 34% vs 100%; HR, 3.78; P = .003), but there was no significant effect of either conditioning regimen or donor source on outcome. Registered at ISRCTN (http://www.isrctn.com/ISRCTN55675535).


Sujet(s)
Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/thérapie , Maladie résiduelle , Protéines nucléaires/génétique , Adolescent , Adulte , Sujet âgé , Femelle , Transplantation de cellules souches hématopoïétiques/mortalité , Humains , Leucémie aigüe myéloïde/mortalité , Mâle , Adulte d'âge moyen , Récidive tumorale locale/génétique , Récidive tumorale locale/mortalité , Maladie résiduelle/diagnostic , Maladie résiduelle/génétique , Nucléophosmine , Récidive , Jeune adulte
18.
Leukemia ; 34(2): 427-440, 2020 02.
Article de Anglais | MEDLINE | ID: mdl-31611628

RÉSUMÉ

Inappropriate localization of proteins can interfere with normal cellular function and drive tumor development. To understand how this contributes to the development of acute myeloid leukemia (AML), we compared the nuclear proteome and transcriptome of AML blasts with normal human CD34+ cells. Analysis of the proteome identified networks and processes that significantly affected transcription regulation including misexpression of 11 transcription factors with seven proteins not previously implicated in AML. Transcriptome analysis identified changes in 40 transcription factors but none of these were predictive of changes at the protein level. The highest differentially expressed protein in AML nuclei compared with normal CD34+ nuclei (not previously implicated in AML) was S100A4. In an extended cohort, we found that over-expression of nuclear S100A4 was highly prevalent in AML (83%; 20/24 AML patients). Knock down of S100A4 in AML cell lines strongly impacted their survival whilst normal hemopoietic stem progenitor cells were unaffected. These data are the first analysis of the nuclear proteome in AML and have identified changes in transcription factor expression or regulation of transcription that would not have been seen at the mRNA level. These data also suggest that S100A4 is essential for AML survival and could be a therapeutic target in AML.


Sujet(s)
Noyau de la cellule/génétique , Leucémie aigüe myéloïde/génétique , Protéome/génétique , Protéine S100A4 liant le calcium/génétique , Transcriptome/génétique , Adolescent , Adulte , Sujet âgé , Antigènes CD34/génétique , Prolifération cellulaire/génétique , Cellules cultivées , Femelle , Humains , Mâle , Adulte d'âge moyen , Cellules souches tumorales/anatomopathologie , Protéomique/méthodes
19.
Blood Adv ; 3(20): 3052-3061, 2019 10 22.
Article de Anglais | MEDLINE | ID: mdl-31648336

RÉSUMÉ

Fms-like tyrosine kinase 3 (Flt3) is expressed on progenitor cells and acute myeloid leukemia (AML) blasts. Fms-like tyrosine kinase 3 ligand (Flt3L) is detectable during homeostasis and increases in hypoplasia due to genetic defects or treatment with cytoreductive agents. Conversely, Flt3+ AML is associated with depletion of Flt3L to undetectable levels. After induction chemotherapy, Flt3L is restored in patients entering complete remission (CR) but remains depressed in those with refractory disease. Weekly sampling reveals marked differences in the kinetics of Flt3L response during the first 6 weeks of treatment, proportionate to the clearance of blasts and cellularity of the bone marrow. In the UK NCRI AML17 trial, Flt3L was measured at day 26 in a subgroup of 140 patients with Flt3 mutation randomized to the tyrosine kinase inhibitor lestaurtinib or placebo. In these patients, attainment of CR was associated with higher Flt3L at day 26 (Mann-Whitney UP < .0001). Day 26 Flt3L was also associated with survival; Flt3L ≤291 pg/mL was associated with inferior event-free survival (EFS), and Flt3L >1185 pg/mL was associated with higher overall survival (OS; P = .0119). The separation of EFS and OS curves increased when minimal residual disease (MRD) status was combined with Flt3L measurement, and Flt3L retained a near-significant association with survival after adjusting for MRD in a proportional hazards model. Serial measurement of Flt3L in patients who had received a hematopoietic stem cell transplant for AML illustrates the potential value of monitoring Flt3L to identify relapse. Measurement of Flt3L is a noninvasive test with the potential to inform clinical decisions in patients with AML.


Sujet(s)
Marqueurs biologiques , Leucémie aigüe myéloïde/sang , Leucémie aigüe myéloïde/diagnostic , Protéines membranaires/sang , Cellules souches tumorales/métabolisme , Expression des gènes , Transplantation de cellules souches hématopoïétiques , Humains , Immunophénotypage , Chimiothérapie d'induction , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/mortalité , Pronostic , Modèles des risques proportionnels , Résultat thérapeutique
20.
Mol Psychiatry ; 24(3): 328-337, 2019 03.
Article de Anglais | MEDLINE | ID: mdl-30647433

RÉSUMÉ

Individuals of African ancestry in the United States and Europe are at increased risk of developing schizophrenia and have poorer clinical outcomes. The antipsychotic clozapine, the only licensed medication for treatment-resistant schizophrenia, is under-prescribed and has high rates of discontinuation in individuals of African ancestry, due in part to increased rates of neutropenia. The genetic basis of lower neutrophil levels in those of African ancestry has not previously been investigated in the context of clozapine treatment. We sought to identify risk alleles in the first genome-wide association study of neutrophil levels during clozapine treatment, in 552 individuals with treatment-resistant schizophrenia and robustly inferred African genetic ancestry. Two genome-wide significant loci were associated with low neutrophil counts during clozapine treatment. The most significantly associated locus was driven by rs2814778 (ß = -0.9, P = 4.21 × 10-21), a known regulatory variant in the atypical chemokine receptor 1 (ACKR1) gene. Individuals homozygous for the C allele at rs2814778 were significantly more likely to develop neutropenia and have to stop clozapine treatment (OR = 20.4, P = 3.44 × 10-7). This genotype, also termed "Duffy-null", has previously been shown to be associated with lower neutrophil levels in those of African ancestry. Our results indicate the relevance of the rs2814778 genotype for those taking clozapine and its potential as a pharmacogenetic test, dependent on the outcome of additional safety studies, to assist decision making in the initiation and on-going management of clozapine treatment.


Sujet(s)
Clozapine/effets indésirables , Neutropénie/induit chimiquement , Neutropénie/génétique , Allèles , Neuroleptiques/usage thérapeutique , /génétique , Clozapine/administration et posologie , Système Duffy/génétique , Femelle , Prédisposition génétique à une maladie , Étude d'association pangénomique/méthodes , Humains , Mâle , Neutropénie/sang , Granulocytes neutrophiles/effets des médicaments et des substances chimiques , Granulocytes neutrophiles/métabolisme , Granulocytes neutrophiles/anatomopathologie , Polymorphisme de nucléotide simple , Récepteurs de surface cellulaire/génétique , Facteurs de risque , Schizophrénie/génétique
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE