Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
J Clin Med ; 13(2)2024 Jan 18.
Article in English | MEDLINE | ID: mdl-38256681

ABSTRACT

Background. Primary resistance of acute myeloid leukemia (AML) to the conventional 3 + 7 intensive chemotherapy and relapses after first-line chemotherapy are two highly challenging clinical scenarios. In these cases, when allogeneic stem cell transplantation is feasible, patients are usually retreated with other chemotherapeutic regimens, as transplantation is still considered, nowadays, the only curative option. Methods. We discuss the mechanisms behind resistance to chemotherapy and offer a comprehensive review on current treatments of refractory/relapsed AML with a focus on novel approaches incorporating the BCL-2 inhibitor venetoclax. Results. Alas, complete remission rates after salvage chemotherapy remain relatively low, between 30 and 60% at best. More recently, the BCL-2 inhibitor venetoclax was combined either with hypomethylating agents or chemotherapy in refractory/relapsed patients. In particular, its combination with chemotherapy offered promising results by achieving higher rates of remission and bridging a substantial number of patients to transplantation. Conclusions. Venetoclax-based approaches might become, in the near future, the new standard of care for refractory/relapsed AML.

2.
Biomedicines ; 11(4)2023 Apr 11.
Article in English | MEDLINE | ID: mdl-37189770

ABSTRACT

TP53 mutations are less frequent in myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) than in solid tumors, except in secondary and therapy-related MDS/AMLs, and in cases with complex monosomal karyotype. As in solid tumors, missense mutations predominate, with the same hotspot mutated codons (particularly codons 175, 248, 273). As TP53-mutated MDS/AMLs are generally associated with complex chromosomal abnormalities, it is not always clear when TP53 mutations occur in the pathophysiological process. It is also uncertain in these MDS/AML cases, which often have inactivation of both TP53 alleles, if the missense mutation is only deleterious through the absence of a functional p53 protein, or through a potential dominant-negative effect, or finally a gain-of-function effect of mutant p53, as demonstrated in some solid tumors. Understanding when TP53 mutations occur in the disease course and how they are deleterious would help to design new treatments for those patients who generally show poor response to all therapeutic approaches.

4.
Haematologica ; 106(12): 3100-3106, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34047178

ABSTRACT

The aim of this study was to characterize a large series of 154 patients with acute promyelocytic leukemia (median age, 53 years; range, 18-90 years) and evaluate real-life outcome after up-front treatment with arsenic trioxide and all-trans retinoic acid. All patients were included in the prospective NAPOLEON registry (NCT02192619) between 2013 and 2019. The acute promyelocytic leukemia was de novo in 91% (n=140) and therapy-related in 9% (n=14); 13% (n=20) of the patients were older than 70 years. At diagnosis bleeding/hemorrhage was present in 38% and thrombosis in 3%. Complete remission was achieved in 152 patients (99%), whereas two patients (1%) experienced induction death within 18 days after starting therapy. With a median follow-up of 1.99 years (95% confidence interval: 1.61-2.30 years) 1-year and 2-year overall survival rates were 97% (95% confidence interval: 94-100%) and 95% (95% confidence interval: 91-99%), respectively. Age above 70 years was associated with a significantly shorter overall survival (P<0.001) compared to that of younger patients. So far no relapses have been observed. Six patients (4%) died in complete remission at a median of 0.95 years after diagnosis (range, 0.18-2.38 years). Our data confirm the efficiency and durability of arsenic trioxide and all-trans retinoic acid therapy in the primary management of adults with low-/intermediate-risk acute promyelocytic leukemia in the real-life setting, irrespective of age.


Subject(s)
Arsenic Trioxide , Leukemia, Promyelocytic, Acute , Adolescent , Adult , Aged , Aged, 80 and over , Arsenic Trioxide/therapeutic use , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Middle Aged , Prospective Studies , Remission Induction , Risk Assessment , Treatment Outcome , Tretinoin/therapeutic use , Young Adult
6.
JCO Glob Oncol ; 7: 455-463, 2021 04.
Article in English | MEDLINE | ID: mdl-33822643

ABSTRACT

The speed and spread of the COVID-19 pandemic has been affecting the entire world for the past several months. OncoAlert is a social media network made up of more than 140 oncology stakeholders: oncologists (medical, radiation, and surgical), oncology nurses, and patient advocates who share the mission of fighting cancer by means of education and dissemination of information. As a response to the COVID-19 pandemic, OncoAlert hosted The Round Table Discussions. We have documented this effort along with further discussion about the COVID-19 pandemic and the consequences on patients living with cancer to disseminate this information to our colleagues worldwide.


Subject(s)
COVID-19/prevention & control , Information Dissemination/methods , Medical Oncology/methods , Neoplasms/therapy , Social Media , Telemedicine/methods , COVID-19/epidemiology , COVID-19/virology , Communicable Disease Control/methods , Epidemics , Global Health/statistics & numerical data , Health Personnel/statistics & numerical data , Humans , Neoplasms/diagnosis , Oncologists/statistics & numerical data , Oncology Nursing/statistics & numerical data , Practice Guidelines as Topic , Public Health/methods , Public Health/statistics & numerical data , SARS-CoV-2/physiology
7.
Methods Mol Biol ; 2267: 165-179, 2021.
Article in English | MEDLINE | ID: mdl-33786791

ABSTRACT

In this chapter, four methods are described that can be used to assess cell cycle status in flow cytometry. The first method is based on the simultaneous analysis of cellular DNA content using a fluorescent DNA dye (propidium iodide) and of a nuclear proliferation marker (Ki-67). The second is based on the differential staining of DNA and RNA using Hoechst 33342 and Pyronin Y: this method is particularly useful to distinguish quiescent cells in G0 phase from G1 cells. Finally, two methods are described based on DNA incorporation of the synthetic nucleosides BrdU and EdU.


Subject(s)
Cell Cycle , Flow Cytometry/methods , Animals , Benzimidazoles/chemistry , Bromodeoxyuridine/chemistry , Cell Line , Fluorescent Dyes/chemistry , Humans , Ki-67 Antigen/metabolism , Pyronine/chemistry
8.
J Clin Oncol ; 39(14): 1575-1583, 2021 05 10.
Article in English | MEDLINE | ID: mdl-33600210

ABSTRACT

PURPOSE: TP53-mutated (TP53m) myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) have very poor outcome irrespective of the treatment received, including 40% responses (20% complete remission [CR]) with azacitidine (AZA) alone, short response duration, and a median overall survival (OS) of approximately 6 months. Eprenetapopt (APR-246), a novel first-in-class drug, leads to p53 protein reconformation and reactivates its proapoptotic and cell-cycle arrest functions. PATIENTS AND METHODS: This phase II study assessed the safety and efficacy of eprenetapopt in combination with AZA in untreated high or very high International Prognostic Scoring System-R TP53m MDS and AML patients. RESULTS: Fifty-two TP53m patients (34 MDS, 18 AML [including seven with more than 30% blasts]) were enrolled. In MDS, we observed an overall response rate (ORR) of 62%, including 47% CR, with a median duration of response at 10.4 months. In AML, the ORR was 33% including 17% CR (27% and 0% CR in AML with less than and more than 30% marrow blasts, respectively). Seventy-three percent of responders achieved TP53 next-generation sequencing negativity (ie, variant allele frequency < 5%). The main treatment-related adverse events were febrile neutropenia (36%) and neurologic adverse events (40%), the latter correlating with a lower glomerular filtration rate at treatment onset (P < .01) and higher age (P = .05), and resolving with temporary drug interruption without recurrence after adequate eprenetapopt dose reduction. With a median follow-up of 9.7 months, median OS was 12.1 months in MDS, and 13.9 and 3.0 months in AML with less than and more than 30% marrow blasts, respectively. CONCLUSION: In this very high-risk population of TP53m MDS and AML patients, eprenetapopt combined with AZA was safe and showed potentially higher ORR and CR rate, and longer OS than reported with AZA alone.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Azacitidine/administration & dosage , Leukemia, Myeloid, Acute/drug therapy , Mutation , Myelodysplastic Syndromes/drug therapy , Quinuclidines/administration & dosage , Tumor Suppressor Protein p53/genetics , Adult , Aged , Aged, 80 and over , Azacitidine/adverse effects , Humans , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/mortality , Middle Aged , Myelodysplastic Syndromes/genetics , Myelodysplastic Syndromes/mortality , Quinuclidines/adverse effects
9.
Blood Adv ; 5(1): 176-184, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33570629

ABSTRACT

CPX-351 is a liposomal formulation of cytarabine and daunorubicin approved for the treatment of adults with newly diagnosed, therapy-related acute myeloid leukemia (t-AML) or AML with myelodysplasia-related changes (MRC-AML). We retrospectively analyzed the efficacy and safety of CPX-351 in a real-world setting in 103 patients from 12 French centers, including the evaluation of molecular abnormalities at baseline and minimal residual disease (MRD) in responding patients, compared with a historical data set from Bordeaux-Toulouse DATAML registry. A favorable safety profile was observed, with a low frequency of alopecia (11%) and gastrointestinal toxicity (50%). The overall response rate after induction was 59%, and MRD <10-3 was achieved in 57% of complete response (CR)/CR with incomplete hematological recovery (CRi) patients. Only the presence of mutated TP53 (P = .02) or PTPN11 (P = .004) predicted lower response in multivariate analysis. Interestingly, high-risk molecular prognosis subgroups defined by 2017 European LeukemiaNet risk stratification, including ASXL1 and RUNX1 mutations, were not associated with a significantly lower response rate using CPX-351. With a median follow-up of 8.6 months, median overall survival (OS) was 16.1 months. Thirty-six patients underwent allogeneic stem cell transplantation with a significantly longer median OS compared with nontransplanted patients (P < .001). In multivariate analyses, only spliceosome mutations were associated with better OS (P = .04). In comparison with intensive chemotherapy, there was no difference in OS for patients <60 years. These data confirm the efficacy and safety of CPX-351 in high-risk AML (t-AML and MRC-AML) in a real-life setting. CPX-351 is a treatment of choice for patients aged ≥60 years.


Subject(s)
Cytarabine , Leukemia, Myeloid, Acute , Adult , Aged , Daunorubicin , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Retrospective Studies
10.
Leukemia ; 35(3): 712-723, 2021 03.
Article in English | MEDLINE | ID: mdl-32581253

ABSTRACT

Intra-tumor heterogeneity portends poor outcome in many cancers. In AML, a higher number of drivers worsens prognosis. The Shannon Index is a robust metric of clonal heterogeneity that accounts for the number of clones, but also their relative abundance. We show that a Shannon Index can be estimated from bulk sequencing, which is correlated (ρ = 0.76) with clonal diversity from single-colony genotyping. In a discovery cohort of 292 patients with sequencing of 43 genes, a higher number of drivers (HR = 1.18, P = 0.028) and a lower Shannon Index (HR = 0.68, P = 0.048), the latter reflecting clonal dominance, are independently associated with worse OS independently of European LeukemiaNet 2017 risk. These findings are validated in an independent cohort of 1184 patients with 111-gene sequencing (number of drivers HR = 1.16, P = 1 × 10-5, Shannon Index HR = 0.81, P = 0.007). By re-interrogating paired diagnosis/relapse exomes from 50 cytogenetically normal AMLs, we find clonal dominance at diagnosis to be correlated with the gain of a significantly higher number of mutations at relapse (P = 6 × 10-6), hence with clonal sweeping. Our results suggest that clonal dominance at diagnosis is associated with the presence of a leukemic phenotype allowing rapid expansion of new clones and driving relapse after chemotherapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Clonal Evolution , Gene Expression Regulation, Neoplastic , Leukemia, Myeloid, Acute/pathology , Female , Follow-Up Studies , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Rate
11.
Leukemia ; 34(9): 2333-2341, 2020 09.
Article in English | MEDLINE | ID: mdl-32076120

ABSTRACT

Data on outcome in older (≥70 years) patients with acute promyelocytic leukemia after treatment with arsenic trioxide (ATO) compared with standard chemotherapy (CTX) is scarce. We evaluated 433 patients (median age, 73.4 years) treated either with ATO+ all-trans retinoic acid (ATO/ATRA; n = 26), CTX/ATRA + ATO during consolidation (CTX/ATRA/ATO; n = 148), or with CTX/ATRA (n = 259). Median follow-up for overall survival (OS) was 4.8 years. Complete remissions (CR) were achieved in 92% with ATO/ATRA and 82% with CTX/ATRA; induction death rates were 8% and 18%, respectively. For analysis of postremission outcomes we combined the ATO/ATRA and CTX/ATRA/ATO groups (ATO/ATRA ± CTX). Cumulative incidence of relapse (CIR) was significantly lower after ATO/ATRA ± CTX compared with CTX/ATRA (P < 0.001). The same held true when restricting the analysis according to the treatment period after the year 2000. OS of patients in CR1 was not different between ATO/ATRA ± CTX compared with CTX/ATRA (P = 0.20). High (>10 × 109/l) white blood cell (WBC) counts at diagnosis were associated with higher CIR (P < 0.001) compared with lower WBC in the CTX/ATRA group, but not in the ATO/ATRA ± CTX group (P = 0.48). ATO, when added to ATRA or CTX/ATRA is feasible and effective in elderly patients for remission induction and consolidation, particularly in patients with high WBC at diagnosis.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Arsenic Trioxide/therapeutic use , Leukemia, Myeloid, Acute/drug therapy , Aged , Aged, 80 and over , Arsenic Trioxide/administration & dosage , Female , Humans , International Cooperation , Leukemia, Myeloid, Acute/genetics , Male , Remission Induction , Treatment Outcome
12.
Haematologica ; 105(6): 1539-1551, 2020 06.
Article in English | MEDLINE | ID: mdl-31488557

ABSTRACT

Myelodysplastic syndromes and acute myeloid leukemia with TP53 mutations are characterized by frequent relapses, poor or short responses, and poor survival with the currently available therapies including chemotherapy and 5-azacitidine (AZA). PRIMA-1Met(APR-246,APR) is a methylated derivative of PRIMA-1, which induces apoptosis in human tumor cells through restoration of the transcriptional transactivation function of mutant p53. Here we show that low doses of APR on its own or in combination with AZA reactivate the p53 pathway and induce an apoptosis program. Functionally, we demonstrate that APR exerts these activities on its own and that it synergizes with AZA in TP53-mutated myelodysplastic syndromes (MDS)/acute myeloid leukemia (AML) cell lines and in TP53-mutated primary cells from MDS/AML patients. Low doses of APR on its own or in combination with AZA also show significant efficacy in vivo Lastly, using transcriptomic analysis, we found that the APR + AZA synergy was mediated by downregulation of the FLT3 pathway in drug-treated cells. Activation of the FLT3 pathway by FLT3 ligand reversed the inhibition of cell proliferation by APR + AZA. These data suggest that TP53-mutated MDS/AML may be better targeted by the addition of APR-246 to conventional treatments.


Subject(s)
Leukemia, Myeloid, Acute , Myelodysplastic Syndromes , Aza Compounds , Azacitidine/pharmacology , Bridged Bicyclo Compounds, Heterocyclic , Humans , Leukemia, Myeloid, Acute/drug therapy , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/drug therapy , Myelodysplastic Syndromes/genetics , Quinuclidines , Tumor Suppressor Protein p53/genetics
13.
Blood ; 134(17): 1441-1444, 2019 10 24.
Article in English | MEDLINE | ID: mdl-31484648

ABSTRACT

Germline DDX41 mutations are involved in familial myelodysplastic syndromes (MDSs) and acute myeloid leukemias (AMLs). We analyzed the prevalence and characteristics of DDX41-related myeloid malignancies in an unselected cohort of 1385 patients with MDS or AML. Using targeted next-generation sequencing, we identified 28 different germline DDX41 variants in 43 unrelated patients, which we classified as causal (n = 21) or unknown significance (n = 7) variants. We focused on the 33 patients having causal variants, representing 2.4% of our cohort. The median age was 69 years; most patients were men (79%). Only 9 patients (27%) had a family history of hematological malignancy, and 15 (46%) had a personal history of cytopenia years before MDS/AML diagnosis. Most patients had a normal karyotype (85%), and the most frequent somatic alteration was a second DDX41 mutation (79%). High-risk DDX41 MDS/AML patients treated with intensive chemotherapy (n = 9) or azacitidine (n = 11) had an overall response rate of 100% or 73%, respectively, with a median overall survival of 5.2 years. Our study highlights that germline DDX41 mutations are relatively common in adult MDS/AML, often without known family history, arguing for systematic screening. Salient features of DDX41-related myeloid malignancies include male preponderance, frequent preexisting cytopenia, additional somatic DDX41 mutation, and relatively good outcome.


Subject(s)
DEAD-box RNA Helicases/genetics , Leukemia, Myeloid, Acute/genetics , Myelodysplastic Syndromes/genetics , Adult , Aged , Aged, 80 and over , Cohort Studies , Female , Germ-Line Mutation , Humans , Male , Middle Aged
14.
Blood ; 133(13): 1495-1506, 2019 03 28.
Article in English | MEDLINE | ID: mdl-30674471

ABSTRACT

Acute promyelocytic leukemia (APL) is often associated with activating FLT3 signaling mutations. These are highly related to hyperleukocytosis, a major adverse risk factor with chemotherapy-based regimens. APL is a model for oncogene-targeted therapies: all-trans retinoic acid (ATRA) and arsenic both target and degrade its ProMyelocytic Leukemia/Retinoic Acid Receptor α (PML/RARA) driver. The combined ATRA/arsenic regimen now cures virtually all patients with standard-risk APL. Although FLT3-internal tandem duplication (ITD) was an adverse risk factor for historical ATRA/chemotherapy regimens, the molecular bases for this effect remain unknown. Using mouse APL models, we unexpectedly demonstrate that FLT3-ITD severely blunts ATRA response. Remarkably, although the transcriptional output of initial ATRA response is unaffected, ATRA-induced PML/RARA degradation is blunted, as is PML nuclear body reformation and activation of P53 signaling. Critically, the combination of ATRA and arsenic fully rescues therapeutic response in FLT3-ITD APLs, restoring PML/RARA degradation, PML nuclear body reformation, P53 activation, and APL eradication. Moreover, arsenic targeting of normal PML also contributes to APL response in vivo. These unexpected results explain the less favorable outcome of FLT3-ITD APLs with ATRA-based regimens, and stress the key role of PML nuclear bodies in APL eradication by the ATRA/arsenic combination.


Subject(s)
Antineoplastic Agents/therapeutic use , Arsenic/therapeutic use , Leukemia, Promyelocytic, Acute/drug therapy , Tretinoin/therapeutic use , fms-Like Tyrosine Kinase 3/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Female , Leukemia, Promyelocytic, Acute/genetics , Mice, Inbred C57BL , Mutation
16.
Expert Rev Hematol ; 12(1): 61-70, 2019 01.
Article in English | MEDLINE | ID: mdl-30334467

ABSTRACT

INTRODUCTION: Myelodysplastic syndromes (MDS) are clonal stem cell disorders mostly affecting the elderly. They are classified into lower and higher risk MDS according to prognostic scoring systems. In higher risk patients, treatments should aim to modify the disease course by avoiding progression to acute myeloid leukemia and, therefore, to improve survival. Areas covered: Stem cell transplantation remains the only curative treatment when feasible, but this concerns a small minority of patients. Treatment is principally based on hypomethylating agents (HMAs). Our understanding of MDS biology has led to the development of drugs targeting key cellular processes such as apoptosis or posttranslational protein changes, microenvironment-like immunotherapy, and gene mutations. Currently, new drugs are mainly being tested in combination with HMAs in several clinical trials. Expert commentary: Significant advances have been made in the field of MDS, especially in molecular typing, which are improving our ability to offer patients risk-adapted therapies. The current challenge in the management of higher risk MDS is to improve outcome by combining classical HMAs with novel drugs.


Subject(s)
Myelodysplastic Syndromes/therapy , Aged , Humans , Risk Factors
SELECTION OF CITATIONS
SEARCH DETAIL
...