Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 4.419
Filtrer
2.
Clin Epigenetics ; 16(1): 63, 2024 May 09.
Article de Anglais | MEDLINE | ID: mdl-38725010

RÉSUMÉ

BACKGROUND: Decitabine (DAC), a DNA methyltransferase inhibitor, has shown efficacy combined with chemotherapy for relapsed or refractory (R/R) acute myeloid leukemia (AML) in adults, but less is known about its efficacy in children. Accordingly, we conducted a study which involved a priming regimen consisting of DAC with cladribine, cytarabine, and granulocyte-stimulating factor (DAC-CLAG) and compared the efficacy and safety of this regimen with CLAG alone. METHODS: A total of 39 R/R AML children who received the CLAG or DAC-CLAG regimen in Shanghai Children's Hospital were retrospectively enrolled in this non-randomized study. These regimens were studied sequentially over time. Twenty-two patients received CLAG from 2015, while 17 patients were administered epigenetic priming with DAC before CLAG from 2020. Patients were subsequently bridged to stem cell transplantation (SCT) or consolidation chemotherapy. Complete remission (CR) and adverse effects were analyzed by Fisher's exact test, and survival was analyzed by the Kaplan-Meier method. RESULTS: DAC-CLAG conferred a numerically higher CR compared to CLAG (70.59% vs 63.64%; P = 0.740). High CR rates occurred in patients with good cytogenetics (P = 0.029) and prior induction without cladribine (P = 0.099). The 1-year event-free survival (EFS) was 64.71% ± 11.59% and 63.31% ± 10.35% in the DAC-CLAG and CLAG group (P = 0.595), and 1-year overall survival (OS) was 81.45% ± 9.72% and 77.01% ± 9.04%, respectively (P = 0.265). The 1-year OS and EFS after SCT were higher in the DAC-CLAG than in the CLAG cohort (100% vs 92.31% ± 7.39%, P = 0.072; 92.31% ± 7.39% vs 85.71% ± 9.35%, P = 0.158). Univariate analysis revealed that a good prognosis included good cytogenetics (P = 0.002), non-complex karyotype (P = 0.056), CR on reinduction (P < 0.0001), and bridging to SCT (P = 0.0007). Use of a hypomethylating agent (P = 0.049) and bridging to SCT (P = 0.011) were independent prognostic factors. Grade 3/4 hematologic toxicity and infection were the main adverse events. CONCLUSIONS: DAC prior to the CLAG regimen improved remission in pediatric R/R AML, and was feasible and well tolerated. CLAG ± DAC as a salvage therapy prior to SCT induced improved survival.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Chloro-2 désoxyadénosine , Cytarabine , Décitabine , Épigenèse génétique , Leucémie aigüe myéloïde , Humains , Décitabine/usage thérapeutique , Décitabine/administration et posologie , Décitabine/pharmacologie , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Mâle , Femelle , Enfant , Enfant d'âge préscolaire , Chloro-2 désoxyadénosine/usage thérapeutique , Chloro-2 désoxyadénosine/administration et posologie , Études rétrospectives , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Adolescent , Épigenèse génétique/effets des médicaments et des substances chimiques , Facteur de stimulation des colonies de granulocytes/administration et posologie , Facteur de stimulation des colonies de granulocytes/usage thérapeutique , Nourrisson , Résultat thérapeutique , Induction de rémission/méthodes
3.
Front Immunol ; 15: 1380451, 2024.
Article de Anglais | MEDLINE | ID: mdl-38765003

RÉSUMÉ

Corticosteroid therapy is the mainstay of immune effector cell-associated neurotoxicity syndrome (ICANS) management, although its use has been associated with worse overall survival (OS) and progression-free survival (PFS) after chimeric antigen receptor T-cell (CAR-T cell) therapy. Many options are being investigated for prophylaxis and management. Accumulating evidence supports the use of intrathecal (IT) chemotherapy for the management of high-grade ICANS. Here, we describe a case of a patient with stage IV Primary mediastinal B-cell lymphoma (PMBCL) successfully treated with IT methotrexate, cytarabine, and dexamethasone as first-line therapy for CD19 CAR-T cell-associated grade IV ICANS. The stable and rapid resolution of ICANS to grade 0 allowed us to discontinue systemic corticosteroid use, avoiding CAR-T cells ablation and ensuring preservation of CAR-T cell function. The described patient achieved a complete radiologic and clinical response to CD19 CAR-T cell therapy and remains disease-free after 9 months. This case demonstrates a promising example of how IT chemotherapy could be used as first-line treatment for the management of high-grade ICANS.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Cytarabine , Dexaméthasone , Injections rachidiennes , Méthotrexate , Humains , Dexaméthasone/administration et posologie , Dexaméthasone/usage thérapeutique , Méthotrexate/administration et posologie , Méthotrexate/usage thérapeutique , Cytarabine/administration et posologie , Cytarabine/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Mâle , Syndromes neurotoxiques/étiologie , Syndromes neurotoxiques/diagnostic , Adulte d'âge moyen , Résultat thérapeutique , Immunothérapie adoptive/effets indésirables , Lymphome B/traitement médicamenteux , Femelle
4.
Hematology ; 29(1): 2343604, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38703055

RÉSUMÉ

PURPOSE: To explore the efficacy and safety of venetoclax-based combination therapy for older patients with newly diagnosed acute myeloid leukemia (AML). METHODS: We performed a systematic review and meta-analysis of clinical trials comparing venetoclax plus hypomethylating agents (HMAs) or low-dose cytarabine (LDAC) with mono-HMAs or LDAC. The random or fixed effects model was applied to the studies based on heterogeneity. Dichotomous data were summarized using the risk ratio (RR) and 95% confidence interval (CI). Continuous variable data were reported as weighted mean differences (WMDs). RESULTS: Nine studies, including a total of 1232 patients, were included in this meta-analysis. Thec complete remission (CR)/complete remission with incomplete hematological recovery (CRi) rate of the venetoclax (Ven) + azacytidine (Aza) group was significantly greater than that of the Aza monotherapy group (RR: 2.42; 95% CI: 1.85-3.15; P < 0.001). Similarly, the CR/CRi rate of the Ven + LDAC group was also significantly greater than that of the LDAC monotherapy group (RR: 2.57; 95% CI: 1.58-4.17; P = 0.00). The same results were observed for OS among these groups. However, the incidence of febrile neutropenia was greater in the Ven + Aza group than in the Ven + Decitabine (Dec) or monotherapy Aza group (RR: 0.69; 95% CI: 0.53-0.90; P = 0.006 and RR: 2.19; 95% CI: 1.58-3.03; P < 0.001, respectively). In addition, the Ven + LDAC group had significantly greater rates of constipation, diarrhea, nausea, and vomiting than the LDAC monotherapy group, with RRs and CIs of 0.61 (95% CI 0.44-0.83, P = 0.002), 1.81 (95% CI 1.22-2.67, P = 0.003), 1.39 (95% CI 1.06-1.82, P = 0.016), and 1.80 (95% CI 1.19-2.72, P = 0.005), respectively. CONCLUSION: Venetoclax combined with azacitidine, decitabine, or LDAC significantly improved the CR/CRi and OS of patients with previously untreated AML. However, venetoclax plus azacitidine or LDAC was more likely to lead to increased febrile neutropenia and gastrointestinal toxicity.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Composés hétérocycliques bicycliques , Leucémie aigüe myéloïde , Sulfonamides , Humains , Composés hétérocycliques bicycliques/usage thérapeutique , Composés hétérocycliques bicycliques/effets indésirables , Composés hétérocycliques bicycliques/administration et posologie , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Sulfonamides/effets indésirables , Leucémie aigüe myéloïde/traitement médicamenteux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Azacitidine/administration et posologie , Azacitidine/effets indésirables , Azacitidine/usage thérapeutique , Résultat thérapeutique , Sujet âgé , Cytarabine/administration et posologie , Cytarabine/usage thérapeutique , Cytarabine/effets indésirables
5.
Nature ; 630(8015): 198-205, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38720074

RÉSUMÉ

Phosphoinositide-3-kinase-γ (PI3Kγ) is implicated as a target to repolarize tumour-associated macrophages and promote antitumour immune responses in solid cancers1-4. However, cancer cell-intrinsic roles of PI3Kγ are unclear. Here, by integrating unbiased genome-wide CRISPR interference screening with functional analyses across acute leukaemias, we define a selective dependency on the PI3Kγ complex in a high-risk subset that includes myeloid, lymphoid and dendritic lineages. This dependency is characterized by innate inflammatory signalling and activation of phosphoinositide 3-kinase regulatory subunit 5 (PIK3R5), which encodes a regulatory subunit of PI3Kγ5 and stabilizes the active enzymatic complex. We identify p21 (RAC1)-activated kinase 1 (PAK1) as a noncanonical substrate of PI3Kγ that mediates this cell-intrinsic dependency and find that dephosphorylation of PAK1 by PI3Kγ inhibition impairs mitochondrial oxidative phosphorylation. Treatment with the selective PI3Kγ inhibitor eganelisib is effective in leukaemias with activated PIK3R5. In addition, the combination of eganelisib and cytarabine prolongs survival over either agent alone, even in patient-derived leukaemia xenografts with low baseline PIK3R5 expression, as residual leukaemia cells after cytarabine treatment have elevated G protein-coupled purinergic receptor activity and PAK1 phosphorylation. Together, our study reveals a targetable dependency on PI3Kγ-PAK1 signalling that is amenable to near-term evaluation in patients with acute leukaemia.


Sujet(s)
Phosphatidylinositol 3-kinases de classe Ib , Leucémies , Transduction du signal , p21-Activated Kinases , Animaux , Humains , Souris , Lignée cellulaire , Phosphatidylinositol 3-kinases de classe Ib/génétique , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Cytarabine/pharmacologie , Cytarabine/usage thérapeutique , Leucémies/traitement médicamenteux , Leucémies/enzymologie , Leucémies/génétique , Leucémies/métabolisme , Mitochondries/effets des médicaments et des substances chimiques , Mitochondries/métabolisme , Phosphorylation oxydative/effets des médicaments et des substances chimiques , p21-Activated Kinases/antagonistes et inhibiteurs , p21-Activated Kinases/métabolisme , Phosphorylation , Tests d'activité antitumorale sur modèle de xénogreffe
6.
J Vet Intern Med ; 38(3): 1618-1625, 2024.
Article de Anglais | MEDLINE | ID: mdl-38700360

RÉSUMÉ

BACKGROUND: Treatment options available for meningoencephalitis of unknown origin (MUO) in dogs are suboptimal, and currently, no single treatment protocol appears to be superior. OBJECTIVES: Compare neurological deterioration rates at 7 days between dogs with MUO treated with corticosteroids alone or combined with cytosine arabinoside (CA) continuous rate infusion (CRI) and compare clinical deterioration and survival at 30 and 100 days. ANIMALS: Sixty-nine dogs with magnetic resonance imaging (MRI) and cerebrospinal fluid (CSF) features or both compatible with MUO. METHODS: Parallel, blinded, randomized controlled trial. Simple randomization into 2 treatment groups: 4 mg/kg/day prednisolone (or dexamethasone equivalent) for 2 days or 200 mg/m2 CA CRI over 8 hours plus 2 mg/kg/day prednisolone. Blinding of the treatment protocol was carried out using reversible redaction of clinical records, and treatment failure was defined as deterioration of neurological assessment or death. Using intention-to-treat analysis, proportions failing treatment at 7, 30, and 100 days were compared using Fisher's exact test. All-cause mortality at 100 days was compared using Kaplan-Meier survival curves. RESULTS: Thirty-five dogs were allocated to corticosteroid only, and 34 dogs were allocated to combined CA CRI and corticosteroid. Proportions failing treatment at 7, 30, and 100 days were 7/35 (20%), 9/35 (26%), and 15/35 (43%) in the corticosteroid-only group and 8/34 (24%), 11/34 (32%), and 23/34 (68%) in the corticosteroid and CA CRI group. All-cause mortality at 100 days was not significantly different between groups (P = .62). Clinically relevant treatment-related adverse effects were not observed. CONCLUSIONS AND CLINICAL IMPORTANCE: We found no difference in outcome between corticosteroid monotherapy and combined cytarabine CRI and corticosteroid therapy at 7, 30, and 100 days after diagnosis in dogs with MUO.


Sujet(s)
Cytarabine , Dexaméthasone , Maladies des chiens , Association de médicaments , Méningoencéphalite , Prednisolone , Animaux , Chiens , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie , Maladies des chiens/traitement médicamenteux , Méningoencéphalite/médecine vétérinaire , Méningoencéphalite/traitement médicamenteux , Mâle , Femelle , Association de médicaments/médecine vétérinaire , Prednisolone/usage thérapeutique , Prednisolone/administration et posologie , Dexaméthasone/usage thérapeutique , Dexaméthasone/administration et posologie , Hormones corticosurrénaliennes/usage thérapeutique , Hormones corticosurrénaliennes/administration et posologie , Perfusions veineuses/médecine vétérinaire
7.
JAMA Netw Open ; 7(5): e2411726, 2024 May 01.
Article de Anglais | MEDLINE | ID: mdl-38753328

RÉSUMÉ

Importance: Disparities in outcomes exist between Black and White patients with acute myeloid leukemia (AML), with Black patients experiencing poorer prognosis compared with their White counterparts. Objective: To assess whether varying intensity of induction therapy to treat pediatric AML is associated with reduced disparities in treatment outcome by race. Design, Setting, and Participants: A comparative effectiveness analysis was conducted of 86 Black and 359 White patients with newly diagnosed AML who were enrolled in the AML02 trial from 2002 to 2008 or the AML08 trial from 2008 to 2017. Statistical analysis was conducted from July 2023 through January 2024. Interventions: Patients in AML02 were randomly assigned to receive standard low-dose cytarabine-based induction therapy or augmented high-dose cytarabine-based induction therapy, whereas patients in AML08 received high-dose cytarabine-based therapy. Main Outcomes and Measures: Cytarabine pharmacogenomic 10-single-nucleotide variant (ACS10) scores were evaluated for association with outcome according to race and treatment arm. Results: This analysis included 86 Black patients (mean [SD] age, 8.8 [6.5] years; 54 boys [62.8%]; mean [SD] leukocyte count, 52 600 [74 000] cells/µL) and 359 White patients (mean [SD] age, 9.1 [6.2] years; 189 boys [52.6%]; mean [SD] leukocyte count, 54 500 [91 800] cells/µL); 70 individuals with other or unknown racial and ethnic backgrounds were not included. Among all patients without core binding factor AML who received standard induction therapy, Black patients had significantly worse outcomes compared with White patients (5-year event-free survival rate, 25% [95% CI, 9%-67%] compared with 56% [95% CI, 46%-70%]; P = .03). By contrast, among all patients who received augmented induction therapy, there were no differences in outcome according to race (5-year event-free survival rate, Black patients, 50% [95% CI, 38%-67%]; White patients, 48% [95% CI, 42%-55%]; P = .78). Among patients who received standard induction therapy, those with low ACS10 scores had a significantly worse 5-year event-free survival rate compared with those with high scores (42.4% [95% CI, 25.6%-59.3%] and 70.0% [95% CI, 56.6%-83.1%]; P = .004); however, among patients who received augmented induction therapy, there were no differences in 5-year event-free survival rates according to ACS10 score (low score, 60.6% [95% CI, 50.9%-70.2%] and high score, 54.8% [95% CI, 47.1%-62.5%]; P = .43). Conclusions and Relevance: In this comparative effectiveness study of pediatric patients with AML treated in 2 consecutive clinical trials, Black patients had worse outcomes compared with White patients after treatment with standard induction therapy, but this disparity was eliminated by treatment with augmented induction therapy. When accounting for ACS10 scores, no outcome disparities were seen between Black and White patients. Our results suggest that using pharmacogenomics parameters to tailor induction regimens for both Black and White patients may narrow the racial disparity gap in patients with AML.


Sujet(s)
Cytarabine , Leucémie aigüe myéloïde , , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/génétique , Mâle , Enfant , Femelle , Cytarabine/usage thérapeutique , Résultat thérapeutique , Enfant d'âge préscolaire , /statistiques et données numériques , /génétique , Pharmacogénétique , Adolescent , Antimétabolites antinéoplasiques/usage thérapeutique , /statistiques et données numériques , Chimiothérapie d'induction/méthodes
8.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 41(4): 404-410, 2024 Apr 10.
Article de Chinois | MEDLINE | ID: mdl-38565504

RÉSUMÉ

OBJECTIVE: To explore the genetic background for a patient with refractory myelodysplastic/myeloproliferative neoplasm (MDS/MPN) with co-morbid neutrophilia patient. METHODS: A MDS/MPN patient who was admitted to the First Affiliated Hospital of Nanjing Medical University in May 2021 was selected as the study subject. RNA sequencing was carried out to identify fusion genes in his peripheral blood mononuclear cells. Fusion gene sequence was searched through transcriptome-wide analysis with a STAR-fusion procedure. The novel fusion genes were verified by quantitative real-time PCR and Sanger sequencing. RESULTS: The patient, a 67-year-old male, had progressive thrombocytopenia. Based on the morphological and molecular examinations, he was diagnosed as MDS/MPN with co-morbid neutropenia, and was treated with demethylating agents and Bcl-2 inhibitors. Seventeen months after the diagnosis, he had progressed to AML. A novel fusion gene NCOR1::GLYR1 was identified by RNA-sequencing in his peripheral blood sample, which was verified by quantitative real-time PCR and Sanger sequencing. The patient had attained morphological remission after a DCAG regimen (a combinatory chemotherapy of decitabine, cytarabine, aclarubicin and granulocyte colony-stimulating factors) plus Chidamide treatment. A significant decrease in the NCOR1::GLYR1 expression was revealed by quantitative real-time PCR at post-chemotherapy evaluation. CONCLUSION: NCOR1::GLYR1 gene is considered as the pathogenic factor for the MDS/MPN patient with neutropenia.


Sujet(s)
Leucémie aigüe myéloïde , Syndromes myélodysplasiques , Neutropénie , Mâle , Humains , Sujet âgé , Syndromes myélodysplasiques/génétique , Agranulocytes , Cytarabine/usage thérapeutique , Corépresseur-1 de récepteur nucléaire
9.
Cells ; 13(7)2024 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-38607071

RÉSUMÉ

Adjuvant treatment for Glioblastoma Grade 4 with Temozolomide (TMZ) inevitably fails due to therapeutic resistance, necessitating new approaches. Apoptosis induction in GB cells is inefficient, due to an excess of anti-apoptotic XPO1/Bcl-2-family proteins. We assessed TMZ, Methotrexate (MTX), and Cytarabine (Ara-C) (apoptosis inducers) combined with XPO1/Bcl-2/Mcl-1-inhibitors (apoptosis rescue) in GB cell lines and primary GB stem-like cells (GSCs). Using CellTiter-Glo® and Caspase-3 activity assays, we generated dose-response curves and analyzed the gene and protein regulation of anti-apoptotic proteins via PCR and Western blots. Optimal drug combinations were examined for their impact on the cell cycle and apoptosis induction via FACS analysis, paralleled by the assessment of potential toxicity in healthy mouse brain slices. Ara-C and MTX proved to be 150- to 10,000-fold more potent in inducing apoptosis than TMZ. In response to inhibitors Eltanexor (XPO1; E), Venetoclax (Bcl-2; V), and A1210477 (Mcl-1; A), genes encoding for the corresponding proteins were upregulated in a compensatory manner. TMZ, MTX, and Ara-C combined with E, V, and A evidenced highly lethal effects when combined. As no significant cell death induction in mouse brain slices was observed, we conclude that this drug combination is effective in vitro and expected to have low side effects in vivo.


Sujet(s)
Amides , Antinéoplasiques , Composés hétérocycliques bicycliques , Glioblastome , Pyrimidines , Sulfonamides , Animaux , Souris , Témozolomide/pharmacologie , Glioblastome/traitement médicamenteux , Glioblastome/métabolisme , Méthotrexate/pharmacologie , Méthotrexate/usage thérapeutique , Cytarabine/pharmacologie , Cytarabine/usage thérapeutique , Antinéoplasiques alcoylants/pharmacologie , Lignée cellulaire tumorale , Antinéoplasiques/pharmacologie , Apoptose
10.
Leukemia ; 38(6): 1307-1314, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38678093

RÉSUMÉ

The therapy of relapsed or refractory (r/r) mantle cell lymphoma (MCL) patients remains a major clinical challenge to date. We conducted a randomized, open-label, parallel-group phase-III trial hypothesizing superior efficacy of rituximab, high-dose cytarabine and dexamethasone with bortezomib (R-HAD + B) versus without (R-HAD) in r/r MCL ineligible for or relapsed after autologous stem cell transplant (ASCT). Primary endpoint was time to treatment failure (TTF), secondary endpoints included response rates, progression free survival, overall survival, and safety. In total, 128 of 175 planned patients were randomized to R-HAD + B (n = 64) or R-HAD (n = 64). Median TTF was 12 vs. 2.6 months (p = 0.045, MIPI-adjusted HR 0.69; 95%CI 0.47-1.02). Overall and complete response rates were 63 vs. 45% (p = 0.049) and 42 vs. 19% (p = 0.0062). A significant treatment effect was seen in the subgroup of patients >65 years (aHR 0.48, 0.29-0.79) and without previous ASCT (aHR 0.52, 0.28-0.96). Toxicity was mostly hematological and attributable to the chemotherapeutic backbone. Grade ≥3 leukocytopenia and lymphocytopenia were more common in R-HAD + B without differences in severe infections between both arms. Bortezomib in combination with chemotherapy can be effective in r/r MCL and should be evaluated further as a therapeutic option, especially if therapy with BTK inhibitors is not an option. Trial registration: NCT01449344.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Bortézomib , Cytarabine , Dexaméthasone , Lymphome à cellules du manteau , Récidive tumorale locale , Rituximab , Humains , Bortézomib/administration et posologie , Bortézomib/usage thérapeutique , Lymphome à cellules du manteau/traitement médicamenteux , Lymphome à cellules du manteau/anatomopathologie , Lymphome à cellules du manteau/mortalité , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Femelle , Mâle , Dexaméthasone/administration et posologie , Dexaméthasone/usage thérapeutique , Dexaméthasone/effets indésirables , Sujet âgé , Adulte d'âge moyen , Cytarabine/administration et posologie , Cytarabine/usage thérapeutique , Rituximab/administration et posologie , Rituximab/usage thérapeutique , Récidive tumorale locale/traitement médicamenteux , Récidive tumorale locale/anatomopathologie , Adulte , Résistance aux médicaments antinéoplasiques , Taux de survie , Sujet âgé de 80 ans ou plus , Études de suivi
11.
Lancet Haematol ; 11(5): e324-e335, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38583455

RÉSUMÉ

BACKGROUND: Whether high-dose cytarabine-based salvage chemotherapy, administered to induce complete remission in patients with poor responsive or relapsed acute myeloid leukaemia scheduled for allogeneic haematopoietic stem-cell transplantation (HSCT) after intensive conditioning confers a survival advantage, is unclear. METHODS: To test salvage chemotherapy before allogeneic HSCT, patients aged between 18 and 75 years with non-favourable-risk acute myeloid leukaemia not in complete remission after first induction or untreated first relapse were randomly assigned 1:1 to remission induction with high-dose cytarabine (3 g/m2 intravenously, 1 g/m2 intravenously for patients >60 years or with a substantial comorbidity) twice daily on days 1-3 plus mitoxantrone (10 mg/m2 intravenously) on days 3-5 or immediate allogeneic HSCT for the disease control group. Block randomisation with variable block lengths was used and patients were stratified by age, acute myeloid leukaemia risk, and disease status. The study was open label. The primary endpoint was treatment success, defined as complete remission on day 56 after allogeneic HSCT, with the aim to show non-inferiority for disease control compared with remission induction with a non-inferiority-margin of 5% and one-sided type 1 error of 2·5%. The primary endpoint was analysed in both the intention-to-treat (ITT) population and in the per-protocol population. The trial is completed and was registered at ClinicalTrials.gov, NCT02461537. FINDINGS: 281 patients were enrolled between Sept 17, 2015, and Jan 12, 2022. Of 140 patients randomly assigned to disease control, 135 (96%) proceeded to allogeneic HSCT, 97 (69%) after watchful waiting only. Of 141 patients randomly assigned to remission induction, 134 (95%) received salvage chemotherapy and 128 (91%) patients subsequently proceeded to allogeneic HSCT. In the ITT population, treatment success was observed in 116 (83%) of 140 patients in the disease control group versus 112 (79%) of 141 patients with remission induction (test for non-inferiority, p=0·036). Among per-protocol treated patients, treatment success was observed in 116 (84%) of 138 patients with disease control versus 109 (81%) of 134 patients in the remission induction group (test for non-inferiority, p=0·047). The difference in treatment success between disease control and remission induction was estimated as 3·4% (95% CI -5·8 to 12·6) for the ITT population and 2·7% (-6·3 to 11·8) for the per-protocol population. Fewer patients with disease control compared with remission induction had non-haematological adverse events grade 3 or worse (30 [21%] of 140 patients vs 86 [61%] of 141 patients, χ2 test p<0·0001). Between randomisation and the start of conditioning, with disease control two patients died from progressive acute myeloid leukaemia and zero from treatment-related complications, and with remission induction two patients died from progressive acute myeloid leukaemia and two from treatment-related complications. Between randomisation and allogeneic HSCT, patients with disease control spent a median of 27 days less in hospital than those with remission induction, ie, the median time in hospital was 15 days (range 7-64) versus 42 days (27-121, U test p<0·0001), respectively. INTERPRETATION: Non-inferiority of disease control could not be shown at the 2·5% significance level. The rate of treatment success was also not statistically better for patients with remission induction. Watchful waiting and immediate transplantation could be an alternative for fit patients with poor response or relapsed acute myeloid leukaemia who have a stem cell donor available. More randomised controlled intention-to-transplant trials are needed to define the optimal treatment before transplantation for patients with active acute myeloid leukaemia. FUNDING: DKMS and the Gert and Susanna Mayer Stiftung Foundation.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Leucémie aigüe myéloïde , Induction de rémission , Transplantation homologue , Humains , Transplantation de cellules souches hématopoïétiques/méthodes , Leucémie aigüe myéloïde/thérapie , Leucémie aigüe myéloïde/traitement médicamenteux , Adulte d'âge moyen , Mâle , Femelle , Adulte , Sujet âgé , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie , Jeune adulte , Adolescent , Mitoxantrone/usage thérapeutique , Mitoxantrone/administration et posologie , Thérapie de rattrapage/méthodes , Résultat thérapeutique , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Récidive
12.
Blood Adv ; 8(12): 3189-3199, 2024 Jun 25.
Article de Anglais | MEDLINE | ID: mdl-38598710

RÉSUMÉ

ABSTRACT: Although it is evident that standard-dose whole-brain radiotherapy as consolidation is associated with significant neurotoxicity, the optimal consolidative strategy for primary central nervous system lymphoma (PCNSL) is not defined. We performed a randomized phase 2 clinical trial via the US Alliance cancer cooperative group to compare myeloablative consolidation supported by autologous stem cell transplantation with nonmyeloablative consolidation after induction therapy for PCNSL. To our knowledge, this is the first randomized trial to be initiated that eliminates whole-brain radiotherapy as a consolidative approach in newly diagnosed PCNSL. Patients aged 18 to 75 years were randomly assigned in a 1:1 manner to induction therapy (methotrexate, temozolomide, rituximab, and cytarabine) followed by consolidation with either thiotepa plus carmustine and autologous stem cell rescue vs induction followed by nonmyeloablative, infusional etoposide plus cytarabine. The primary end point was progression-free survival (PFS). A total of 113 patients were randomized, and 108 (54 in each arm) were evaluable. More patients in the nonmyeloablative arm experienced progressive disease or death during induction (28% vs 11%; P = .05). Thirty-six patients received autologous stem cell transplant, and 34 received nonmyeloablative consolidation. The estimated 2-year PFS was higher in the myeloablative vs nonmyeloablative arm (73% vs 51%; P = .02). However, a planned secondary analysis, landmarked at start of the consolidation, revealed that the estimated 2-year PFS in those who completed consolidation therapy was not significantly different between the arms (86% vs 71%; P = .21). Both consolidative strategies yielded encouraging efficacy and similar toxicity profiles. This trial was registered at www.clininicals.gov as #NCT01511562.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du système nerveux central , Lymphomes , Humains , Adulte d'âge moyen , Tumeurs du système nerveux central/thérapie , Tumeurs du système nerveux central/mortalité , Adulte , Femelle , Mâle , Sujet âgé , Lymphomes/thérapie , Lymphomes/mortalité , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Jeune adulte , Transplantation de cellules souches hématopoïétiques/méthodes , Transplantation autologue , Adolescent , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie , Résultat thérapeutique , Chimiothérapie de consolidation , Association thérapeutique
13.
Oncologist ; 29(6): e796-e802, 2024 Jun 03.
Article de Anglais | MEDLINE | ID: mdl-38581718

RÉSUMÉ

BACKGROUND: A consolidation strategy has not been established for transplant-ineligible elderly patients with primary central nervous system lymphoma (PCNSL). In this study, we aimed to retrospectively evaluate the clinical outcomes of etoposide and cytarabine (EA) as consolidation chemotherapy for transplant-ineligible patients with PCNSL following high-dose methotrexate (MTX)-based induction chemotherapy. MATERIALS AND METHODS: Between 2015 and 2021, newly diagnosed transplant-ineligible patients with PCNSL with diffuse large B-cell lymphoma were consecutively enrolled. All enrolled patients were over 60 years old and received EA consolidation after achieving a complete or partial response following induction chemotherapy. RESULTS: Of the 85 patients who achieved a complete or partial response to MTX-based induction chemotherapy, 51 received EA consolidation chemotherapy. Among the 25 (49.0%, 25/51) patients in partial remission before EA consolidation, 56% (n = 14) achieved complete remission after EA consolidation. The median overall survival and progression-free survival were 43 and 13 months, respectively. Hematological toxicities were most common, and all patients experienced grade 4 neutropenia and thrombocytopenia. Forty-eight patients experienced febrile neutropenia during consolidation chemotherapy, and 4 patients died owing to treatment-related complications. CONCLUSION: EA consolidation chemotherapy for transplant-ineligible, elderly patients with PCNSL improved response rates but showed a high relapse rate and short progression-free survival. The incidences of treatment-related mortality caused by hematologic toxicities and severe infections were very high, even after dose modification. Therefore, the use of EA consolidation should be reconsidered in elderly patients with PCNSL.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Tumeurs du système nerveux central , Chimiothérapie de consolidation , Cytarabine , Étoposide , Humains , Étoposide/administration et posologie , Étoposide/usage thérapeutique , Étoposide/effets indésirables , Femelle , Mâle , Cytarabine/administration et posologie , Cytarabine/effets indésirables , Cytarabine/usage thérapeutique , Sujet âgé , Tumeurs du système nerveux central/traitement médicamenteux , Tumeurs du système nerveux central/mortalité , Chimiothérapie de consolidation/méthodes , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Études rétrospectives , Adulte d'âge moyen , Sujet âgé de 80 ans ou plus , Résultat thérapeutique , Lymphome B diffus à grandes cellules/traitement médicamenteux , Lymphome B diffus à grandes cellules/mortalité
14.
Leuk Res ; 140: 107485, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38579483

RÉSUMÉ

Over the years, the overall survival of older patients diagnosed with acute myeloid leukemia (AML) has not significantly increased. Although standard cytotoxic therapies that rapidly eliminate dividing myeloblasts are used to induce remission, relapse can occur due to surviving therapy-resistant leukemic stem cells (LSCs). Hence, anti-LSC strategies have become a key target to cure AML. We have recently shown that previously approved cardiac glycosides and glucocorticoids target LSC-enriched CD34+ cells in the primary human AML 8227 model with more efficacy than normal hematopoietic stem cells (HSCs). To translate these in vitro findings into humans, we developed a mathematical model of stem cell dynamics that describes the stochastic evolution of LSCs in AML post-standard-of-care. To this, we integrated population pharmacokinetic-pharmacodynamic (PKPD) models to investigate the clonal reduction potential of several promising candidate drugs in comparison to cytarabine, which is commonly used in high doses for consolidation therapy in AML patients. Our results suggest that cardiac glycosides (proscillaridin A, digoxin and ouabain) and glucocorticoids (budesonide and mometasone) reduce the expansion of LSCs through a decrease in their viability. While our model predicts that effective doses of cardiac glycosides are potentially too toxic to use in patients, simulations show the possibility of mometasone to prevent relapse through the glucocorticoid's ability to drastically reduce LSC population size. This work therefore highlights the prospect of these treatments for anti-LSC strategies and underlines the use of quantitative approaches to preclinical drug translation in AML.


Sujet(s)
Leucémie aigüe myéloïde , Cellules souches tumorales , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Cellules souches tumorales/effets des médicaments et des substances chimiques , Cellules souches tumorales/anatomopathologie , Modèles théoriques , Cytarabine/usage thérapeutique , Cytarabine/pharmacologie
15.
Ann Hematol ; 103(6): 1967-1977, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38676765

RÉSUMÉ

Acute myeloid leukemia (AML) is a highly heterogeneous hematological malignancy. Cytarabine (Ara-C)-based chemotherapy is the primary treatment for AML, but currently known prognostic risk stratification factors cannot fully explain the individual differences in outcome of patients. In this article, we reported that patients with homozygous GLI1 rs2228224 mutation (AA genotype) had a significantly lower complete remission rate than those with GG wild type (54.17% vs.76.02%, OR = 1.993, 95% CI: 1.062-3.504, P = 0.031). GLI1 rs2229300 T allele carriers had remarkably shorter overall survival (513 vs. 645 days, P = 0.004) and disease-free survival (342 vs. 456 days, P = 0.033) than rs2229300 GG carriers. Rs2229300 G > T variation increased the transcriptional activity of GLI1. CCND1, CD44 and PROM1 were potential target genes differentially regulated by GLI1 rs2229300. Our results demonstrated for the first time that GLI1 polymorphisms influence chemosensitivity and prognosis of young de novo AML patients treated with Ara-C.


Sujet(s)
Cytarabine , Leucémie aigüe myéloïde , Induction de rémission , Protéine à doigt de zinc GLI1 , Humains , Protéine à doigt de zinc GLI1/génétique , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/mortalité , Femelle , Mâle , Adulte , Adolescent , Jeune adulte , Pronostic , Polymorphisme de nucléotide simple , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Survie sans rechute
16.
Blood Cancer J ; 14(1): 54, 2024 Mar 26.
Article de Anglais | MEDLINE | ID: mdl-38531863

RÉSUMÉ

Despite an increasing desire to use historical cohorts as "synthetic" controls for new drug evaluation, limited data exist regarding the comparability of real-world outcomes to those in clinical trials. Governmental cancer data often lacks details on treatment, response, and molecular characterization of disease sub-groups. The Australasian Leukaemia and Lymphoma Group National Blood Cancer Registry (ALLG NBCR) includes source information on morphology, cytogenetics, flow cytometry, and molecular features linked to treatment received (including transplantation), response to treatment, relapse, and survival outcome. Using data from 942 AML patients enrolled between 2012-2018, we assessed age and disease-matched control and interventional populations from published randomized trials that led to the registration of midostaurin, gemtuzumab ozogamicin, CPX-351, oral azacitidine, and venetoclax. Our analyses highlight important differences in real-world outcomes compared to clinical trial populations, including variations in anthracycline type, cytarabine intensity and scheduling during consolidation, and the frequency of allogeneic hematopoietic cell transplantation in first remission. Although real-world outcomes were comparable to some published studies, notable differences were apparent in others. If historical datasets were used to assess the impact of novel therapies, this work underscores the need to assess diverse datasets to enable geographic differences in treatment outcomes to be accounted for.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Leucémie aigüe myéloïde , Humains , Récidive tumorale locale/traitement médicamenteux , Résultat thérapeutique , Cytarabine/usage thérapeutique , Gemtuzumab/usage thérapeutique , Leucémie aigüe myéloïde/thérapie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique
18.
Lancet Haematol ; 11(5): e358-e367, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38555923

RÉSUMÉ

BACKGROUND: Chimeric antigen receptor (CAR) T cells targeting CD30 are safe and have promising activity when preceded by lymphodepleting chemotherapy. We aimed to determine the safety of anti-CD30 CAR T cells as consolidation after autologous haematopoietic stem-cell transplantation (HSCT) in patients with CD30+ lymphoma at high risk of relapse. METHODS: This phase 1 dose-escalation study was performed at two sites in the USA. Patients aged 3 years and older, with classical Hodgkin lymphoma or non-Hodgkin lymphoma with CD30+ disease documented by immunohistochemistry, and a Karnofsky performance score of more than 60% planned for autologous HSCT were eligible if they were considered high risk for relapse as defined by primary refractory disease or relapse within 12 months of initial therapy or extranodal involvement at the start of pre-transplantation salvage therapy. Patients received a single infusion of CAR T cells (2 × 107 CAR T cells per m2, 1 × 108 CAR T cells per m2, or 2 × 108 CAR T cells per m2) as consolidation after trilineage haematopoietic engraftment (defined as absolute neutrophil count ≥500 cells per µL for 3 days, platelet count ≥25 × 109 platelets per L without transfusion for 5 days, and haemoglobin ≥8 g/dL without transfusion for 5 days) following carmustine, etoposide, cytarabine, and melphalan (BEAM) and HSCT. The primary endpoint was the determination of the maximum tolerated dose, which was based on the rate of dose-limiting toxicity in patients who received CAR T-cell infusion. This study is registered with ClinicalTrials.gov (NCT02663297) and enrolment is complete. FINDINGS: Between June 7, 2016, and Nov 30, 2020, 21 patients were enrolled and 18 patients (11 with Hodgkin lymphoma, six with T-cell lymphoma, one with grey zone lymphoma) were infused with anti-CD30 CAR T cells at a median of 22 days (range 16-44) after autologous HSCT. There were no dose-limiting toxicities observed, so the highest dose tested, 2 × 108 CAR T cells per m2, was determined to be the maximum tolerated dose. One patient had grade 1 cytokine release syndrome. The most common grade 3-4 adverse events were lymphopenia (two [11%] of 18) and leukopenia (two [11%] of 18). There were no treatment-related deaths. Two patients developed secondary malignancies approximately 2 years and 2·5 years following treatment (one stage 4 non-small cell lung cancer and one testicular cancer), but these were judged unrelated to treatment. At a median follow-up of 48·2 months (IQR 27·5-60·7) post-infusion, the median progression-free survival for all treated patients (n=18) was 32·3 months (95% CI 4·6 months to not estimable) and the median progression-free survival for treated patients with Hodgkin lymphoma (n=11) has not been reached. The median overall survival for all treated patients has not been reached. INTERPRETATION: Anti-CD30 CAR T-cell infusion as consolidation after BEAM and autologous HSCT is safe, with low rates of toxicity and encouraging preliminary activity in patients with Hodgkin lymphoma at high risk of relapse, highlighting the need for larger studies to confirm these findings. FUNDING: National Heart Lung and Blood Institute, University Cancer Research Fund at the Lineberger Comprehensive Cancer Center.


Sujet(s)
Transplantation de cellules souches hématopoïétiques , Immunothérapie adoptive , Antigènes CD30 , Transplantation autologue , Humains , Transplantation de cellules souches hématopoïétiques/méthodes , Transplantation de cellules souches hématopoïétiques/effets indésirables , Mâle , Femelle , Adulte d'âge moyen , Adulte , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Sujet âgé , Adolescent , Maladie de Hodgkin/thérapie , Maladie de Hodgkin/immunologie , Jeune adulte , Enfant , Récepteurs chimériques pour l'antigène/immunologie , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Melphalan/usage thérapeutique , Melphalan/administration et posologie , Lymphome malin non hodgkinien/thérapie , Lymphome malin non hodgkinien/immunologie , Carmustine/usage thérapeutique , Carmustine/administration et posologie , Étoposide/usage thérapeutique , Étoposide/administration et posologie , Enfant d'âge préscolaire , Cytarabine/usage thérapeutique , Cytarabine/administration et posologie
19.
Blood ; 143(23): 2363-2372, 2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38452207

RÉSUMÉ

ABSTRACT: We previously demonstrated that a reduced-intensity chemotherapy schedule can safely replace hyper-CVAD (cyclophosphamide-vincristine-doxorubicin [Adriamycin]-dexamethasone) cycle 1 when combined with imatinib in adults with Philadelphia-positive acute lymphoblastic leukemia. In the present randomized GRAAPH-2014 trial, we used nilotinib and addressed the omission of cytarabine (Ara-C) in consolidation. The primary objective was the major molecular response (MMR) rate measured by BCR::ABL1 quantification after cycle 4 (end of consolidation). All patients were eligible for allogeneic stem cell transplant (SCT), whereas those in MMR could receive autologous SCT, followed by 2-year imatinib maintenance in both cases. After the enrollment of 156 of 265 planed patients, the data and safety monitoring board decided to hold the randomization because of an excess of relapse in the investigational arm. Among the 155 evaluable patients, 76 received Ara-C during consolidation (arm A) and 79 did not (arm B). Overall, 133 patients (85%) underwent SCT, 93 allogeneic and 40 autologous. The noninferiority end point regarding MMR was reached with 71.1% (arm A) and 77.2% (arm B) of patients reaching MMR. However, the 4-year cumulative incidence of relapse was higher in arm B compared with arm A (31.3% [95% confidence interval {CI}, 21.1%-41.9%] vs 13.2% [95% CI, 6.7%-21.9%]; P = .017), which translated to a lower relapse-free survival. With a median follow-up of 3.8 years, 4-year overall survival was 79.0% (95% CI, 70.6%-89.3%) in arm A vs 73.4% (95% CI, 63.9%-84.4%) in arm B (P = .35). Despite a noninferior rate of MMR, more relapses were observed when ARA-C was omitted without impact on survival. ClinicalTrials.gov ID, NCT02611492.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Cytarabine , Leucémie-lymphome lymphoblastique à précurseurs B et T , Pyrimidines , Humains , Cytarabine/administration et posologie , Cytarabine/usage thérapeutique , Femelle , Mâle , Adulte , Adulte d'âge moyen , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/mortalité , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Pyrimidines/usage thérapeutique , Pyrimidines/administration et posologie , Sujet âgé , Jeune adulte , Adolescent , Protéines de fusion bcr-abl/génétique , Transplantation de cellules souches hématopoïétiques
20.
Am J Hematol ; 99(6): 1180-1183, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38526002
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...