Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 877
Filtrer
1.
J Immunother Cancer ; 12(6)2024 Jun 06.
Article de Anglais | MEDLINE | ID: mdl-38844406

RÉSUMÉ

The bispecific T cell-binding antibody blinatumomab (CD19/CD3) is widely and successfully used for the treatment of children with relapsed or refractory B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Here, we report the efficacy of a single course of blinatumomab instead of consolidation chemotherapy to eliminate minimal residual disease (MRD) and maintain stable MRD-negativity in children with primary BCP-ALL.Between February 2020 and November 2022, 177 children with non-high-risk BCP-ALL were enrolled in the ALL-MB 2019 pilot study (NCT04723342). Patients received the usual risk-adapted induction therapy according to the ALL-MB 2015 protocol. Those who achieved a complete remission at the end of induction (EOI) received treatment with blinatumomab immediately after induction at a dose of 5 µg/m2/day for 7 days and 21 days at a dose of 15 µg/m2/day, followed by 12 months of maintenance therapy. MRD was measured using multicolor flow cytometry (MFC) at the EOI, then immediately after blinatumomab treatment, and then four times during maintenance therapy at 3-month intervals.All 177 patients successfully completed induction therapy and achieved a complete hematological remission. In 174 of these, MFC-MRD was measured at the EOI. 143 patients (82.2%) were MFC-MRD negative and the remaining 31 patients had varying degrees of MFC-MRD positivity.MFC-MRD was assessed in all 176 patients who completed the blinatumomab course. With one exception, all patients achieved MFC-MRD negativity after blinatumomab, regardless of the MFC-MRD score at EOI. One adolescent girl with high MFC-MRD positivity at EOI remained MFC-MRD positive. Of 175 patients who had completed 6 months of maintenance therapy, MFC-MRD data were available for 156 children. Of these, 155 (99.4%) were MFC-MRD negative. Only one boy with t(12;21) (p13;q22)/ETV6::RUNX1 became MFC-MRD positive again. The remaining 174 children had completed the entire therapy. MFC-MRD was examined in 154 of them, and 153 were MFC-MRD negative. A girl with hypodiploid BCP-ALL showed a reappearance of MFC-MRD with subsequent relapse.In summary, a single 28-day course of blinatumomab immediately after induction, followed by 12 months of maintenance therapy, is highly effective in achieving MRD-negativity in children with newly diagnosed non-high risk BCP-ALL and maintaining MRD-negative remission at least during the treatment period.


Sujet(s)
Anticorps bispécifiques , Maladie résiduelle , Leucémie-lymphome lymphoblastique à précurseurs B , Adolescent , Enfant , Enfant d'âge préscolaire , Femelle , Humains , Nourrisson , Mâle , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/pharmacologie , Chimiothérapie de consolidation/méthodes , Chimiothérapie de maintenance/méthodes , Maladie résiduelle/traitement médicamenteux , Projets pilotes , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux
2.
Haematologica ; 109(6): 1668-1676, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38832422

RÉSUMÉ

Immunotherapy has revolutionized treatment for a wide variety of cancers yet its use has been relatively limited in childhood malignancies. With the introduction of bispecific T-cell engagers (BiTE®) and chimeric antigen T-cell receptor technologies, previously refractory patients have attained remission, including molecularly negative states of disease, thus providing the possibility of long-term cure. Blinatumomab is a widely available CD3-CD19 BiTE that has dramatically changed the landscape of therapy for some children with precursor-B acute lymphoblastic leukemias (B-ALL) and lymphoblastic lymphomas. Challenges remain with using BiTE in a broader population although the appeal of now-confirmed reduced toxicity and deeper molecular remissions suggests that this approach will be an essential part of future treatment of childhood B-ALL. Herein, we review some of the pertinent literature covering clinical trials with blinatumomab and address future approaches and combination trials including BiTE.


Sujet(s)
Anticorps bispécifiques , Leucémie-lymphome lymphoblastique à précurseurs B , Lymphocytes T , Humains , Anticorps bispécifiques/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Enfant , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Essais cliniques comme sujet , Antigènes CD19/immunologie , Résultat thérapeutique , Antinéoplasiques immunologiques/usage thérapeutique
3.
Math Biosci Eng ; 21(4): 5164-5180, 2024 Mar 05.
Article de Anglais | MEDLINE | ID: mdl-38872531

RÉSUMÉ

B-cell acute lymphoblastic leukemia (B-ALL) is a malignant blood disorder, particularly detrimental to children and adolescents, with recurrent or unresponsive cases contributing significantly to cancer-associated fatalities. IKBKE, associated with innate immunity, tumor promotion, and drug resistance, remains poorly understood in the context of B-ALL. Thus, this research aimed to explore the impact of the IKBKE inhibitor MCCK1 on B-ALL cells. The study encompassed diverse experiments, including clinical samples, in vitro and in vivo investigations. Quantitative real-time fluorescence PCR and protein blotting revealed heightened IKBKE mRNA and protein expression in B-ALL patients. Subsequent in vitro experiments with B-ALL cell lines demonstrated that MCCK1 treatment resulted in reduced cell viability and survival rates, with flow cytometry indicating cell cycle arrest. In vivo experiments using B-ALL mouse tumor models substantiated MCCK1's efficacy in impeding tumor proliferation. These findings collectively suggest that IKBKE, found to be elevated in B-ALL patients, may serve as a promising drug target, with MCCK1 demonstrating potential for inducing apoptosis in B-ALL cells both in vitro and in vivo.


Sujet(s)
Apoptose , Prolifération cellulaire , I-kappa B Kinase , Animaux , Humains , Souris , I-kappa B Kinase/antagonistes et inhibiteurs , I-kappa B Kinase/métabolisme , Lignée cellulaire tumorale , Apoptose/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des médicaments et des substances chimiques , Femelle , Survie cellulaire/effets des médicaments et des substances chimiques , Mâle , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Enfant , Adolescent , Tests d'activité antitumorale sur modèle de xénogreffe , Inhibiteurs de protéines kinases/pharmacologie
4.
Paediatr Drugs ; 26(4): 459-467, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38780741

RÉSUMÉ

Inotuzumab ozogamicin (BESPONSA™) is a CD22-targeted monoclonal antibody drug conjugate (ADC) developed by Pfizer for the treatment of CD22-postive B-cell precursor acute lymphoblastic leukaemia (ALL). Inotuzumab ozogamicin comprises a humanized IgG4 anti-CD22 monoclonal antibody covalently linked to the potent DNA-binding cytotoxic agent N-acetyl-gamma-calicheamicin dimethylhydrazide (CalichDMH) via a linker. Inotuzumab ozogamicin binds to CD22-expressing tumour cells, facilitating the delivery of conjugated CalichDMH, which after intracellular activation induces double strand DNA breaks, ultimately leading to cell cycle arrest and apoptotic cell death. Inotuzumab ozogamicin is approved in the USA, Europe and several countries worldwide for the treatment of relapsed or refractory CD22-positive B-cell precursor ALL in adults. On 6 March 2024, inotuzumab ozogamicin received its first pediatric approval in the USA for this indication in patients aged ≥ 1 years. Inotuzumab ozogamicin has since been approved in Japan in March 2024 for the same indication in pediatric patients. This article summarizes the milestones in the development of inotuzumab ozogamicin leading to this first approval for the treatment of relapsed or refractory CD22-positive B-cell precursor ALL in pediatric patients.


Sujet(s)
Agrément de médicaments , Inotuzumab ozogamicine , Humains , Enfant , Lectine-2 de type Ig liant l'acide sialique/immunologie , Antinéoplasiques immunologiques/effets indésirables , Antinéoplasiques immunologiques/usage thérapeutique , Antinéoplasiques immunologiques/pharmacocinétique , Antinéoplasiques immunologiques/pharmacologie , Antinéoplasiques immunologiques/administration et posologie , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Anticorps monoclonaux humanisés/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/pharmacologie , Anticorps monoclonaux humanisés/pharmacocinétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux
5.
Ann Hematol ; 103(7): 2541-2543, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38789590

RÉSUMÉ

Blinatumomab as a single agent has demonstrated superiority over salvage chemotherapy in patients with relapsed and refractory B-cell acute lymphoblastic leukemia (B-ALL), with manageable safety and efficacy. Though known to have anticipated drug toxicities including cytokine release syndrome (CRS) and neurotoxicity, there is only one prior report of macrophage activating syndrome (MAS) due to blinatumomab. Case Presentation: We report the first case of blinatumomab-induced MAS in an adult. The patient presented with fever, cough, and weakness on the second cycle of blinatumomab. Complete blood count was notable for severe leukopenia, with comprehensive metabolic panel notable for elevated alkaline phosphatase, AST, ALT, LDH, and hyperferritinemia consistent with MAS. The patient was already in MRD-negative remission at presentation with MAS. She responded rapidly to withholding the drug and administration of both tocilizumab and dexamethasone. She was able to restart therapy with blinatumomab dosed at 9 mcg/day with no recurrence of symptoms. Though MAS is not an expected association with blinatumomab, the risk for CRS is. Secondary MAS in this case likely shares a mechanism with other hyperinflammatory conditions. Management includes holding the offending agent, like blinatumomab, and administering tocilizumab and dexamethasone. Future research will be needed to predict which patients are at highest risk to develop MAS after similar T-cell therapies.


Sujet(s)
Anticorps bispécifiques , Syndrome d'activation macrophagique , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Anticorps bispécifiques/effets indésirables , Anticorps bispécifiques/usage thérapeutique , Femelle , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Syndrome d'activation macrophagique/induit chimiquement , Syndrome d'activation macrophagique/étiologie , Adulte , Dexaméthasone/effets indésirables , Dexaméthasone/usage thérapeutique , Anticorps monoclonaux humanisés/effets indésirables , Anticorps monoclonaux humanisés/usage thérapeutique , Antinéoplasiques/effets indésirables , Antinéoplasiques/usage thérapeutique
6.
BMC Med Genomics ; 17(1): 149, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811988

RÉSUMÉ

Pediatric B-cell acute lymphoblastic leukemia (B-ALL) is a highly heterogeneous disease. According to large-scale RNA sequencing (RNA-seq) data, B-ALL patients can be divided into more than 10 subgroups. However, many genomic defects associated with resistance mechanisms have not yet been identified. As an individual clinical tool for molecular diagnostic risk classification, RNA-seq and gene expression pattern-based therapy could be potential upcoming strategies. In this study, we retrospectively analyzed the RNA-seq gene expression profiles of 45 children whose molecular diagnostic classifications were inconsistent with the response to chemotherapy. The relationship between the transcriptome and chemotherapy response was analyzed. Fusion gene identification was conducted for the included patients who did not have known high-risk associated fusion genes or gene mutations. The most frequently detected fusion gene pair in the high-risk group was the DHRSX duplication, which is a novel finding. Fusions involving ABL1, LMNB2, NFATC1, PAX5, and TTYH3 at onset were more frequently detected in the high-risk group, while fusions involving LFNG, TTYH3, and NFATC1 were frequently detected in the relapse group. According to the pathways involved, the underlying drug resistance mechanism is related to DNA methylation, autophagy, and protein metabolism. Overall, the implementation of an RNA-seq diagnostic system will identify activated markers associated with chemotherapy response, and guide future treatment adjustments.


Sujet(s)
Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Enfant , Mâle , Femelle , Enfant d'âge préscolaire , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/diagnostic , Analyse de séquence d'ARN , Adolescent , Résistance aux médicaments antinéoplasiques/génétique , Nourrisson , Études rétrospectives , Protéines de fusion oncogènes/génétique
7.
J Pediatr Hematol Oncol ; 46(5): e317-e321, 2024 Jul 01.
Article de Anglais | MEDLINE | ID: mdl-38775421

RÉSUMÉ

Thiopurine-methyltransferase (TPMT) and nudix-hydrolase-15 (NUDT15) are enzymes relevant to the metabolism of thiopurine medications, used to treat immunologic disorders and malignancies. Standard dosing administered in the setting of TPMT/NUDT15 dysfunction can cause excessive cytotoxic metabolites and life-threatening complications. We describe an adolescent with high-risk B-cell acute lymphoblastic leukemia (ALL) whose TPMT/NUDT15 status was unknown due to lack of insurance approval for genetic testing. He subsequently developed myelosuppression and severe veno-occlusive disease (VOD) after receiving 6-mercaptopurine (6-MP). Our patient provides an example of a very rare 6-MP-related toxicity and the potential benefit of TPMT/NUDT15 screening before initiating thiopurine therapy.


Sujet(s)
Antimétabolites antinéoplasiques , Maladie veno-occlusive hépatique , Mercaptopurine , Humains , Mercaptopurine/effets indésirables , Mercaptopurine/administration et posologie , Mâle , Maladie veno-occlusive hépatique/induit chimiquement , Maladie veno-occlusive hépatique/anatomopathologie , Adolescent , Antimétabolites antinéoplasiques/effets indésirables , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Methyltransferases/génétique
8.
Nat Commun ; 15(1): 4557, 2024 May 29.
Article de Anglais | MEDLINE | ID: mdl-38811530

RÉSUMÉ

Glucocorticoid (GC) resistance in childhood relapsed B-cell acute lymphoblastic leukemia (B-ALL) represents an important challenge. Despite decades of clinical use, the mechanisms underlying resistance remain poorly understood. Here, we report that in B-ALL, GC paradoxically induce their own resistance by activating a phospholipase C (PLC)-mediated cell survival pathway through the chemokine receptor, CXCR4. We identify PLC as aberrantly activated in GC-resistant B-ALL and its inhibition is able to induce cell death by compromising several transcriptional programs. Mechanistically, dexamethasone (Dex) provokes CXCR4 signaling, resulting in the activation of PLC-dependent Ca2+ and protein kinase C signaling pathways, which curtail anticancer activity. Treatment with a CXCR4 antagonist or a PLC inhibitor improves survival of Dex-treated NSG mice in vivo. CXCR4/PLC axis inhibition significantly reverses Dex resistance in B-ALL cell lines (in vitro and in vivo) and cells from Dex resistant ALL patients. Our study identifies how activation of the PLC signalosome in B-ALL by Dex limits the upfront efficacy of this chemotherapeutic agent.


Sujet(s)
Dexaméthasone , Résistance aux médicaments antinéoplasiques , Glucocorticoïdes , Récepteurs CXCR4 , Transduction du signal , Type C Phospholipases , Récepteurs CXCR4/métabolisme , Récepteurs CXCR4/génétique , Humains , Animaux , Transduction du signal/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/génétique , Dexaméthasone/pharmacologie , Type C Phospholipases/métabolisme , Lignée cellulaire tumorale , Glucocorticoïdes/pharmacologie , Souris , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Souris de lignée NOD , Survie cellulaire/effets des médicaments et des substances chimiques
9.
J Assoc Physicians India ; 72(3): 97-99, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38736127

RÉSUMÉ

We present a case of a 24-year-old female recently diagnosed with acute leukemia who came with complaints of fever for 14 days, progressive lower limb weakness, and multiple episodes of vomiting in the last 1 day. In nerve conduction studies, a diagnosis of Guillain-Barré syndrome (GBS) was established. Fever with thrombocytopenia workup revealed a positive dengue nonstructural protein 1 (NS1) and immunoglobulin M (IgM) report. Immunophenotyping confirmed pre-B acute lymphoblastic leukemia (ALL). As leukemia is an immunocompromised state, the peripheral nervous system vulnerability is increased, or infection could precipitate an immune neuropathy. About 10% of adult ALL presents with central nervous system (CNS) leukemias; a higher incidence is seen in mature B ALL. There is some evidence to suggest immunosuppression secondary to intensive chemotherapy (vincristine-induced dying back neuropathy), which was not started in our case. This rare combination in a short period of time with a worsening situation paralyzed the line of management. Few reports described GBS in patients with dengue in adults. The association of Guillan-Barre syndrome and ALL could be coincidental or has a pathophysiological basis and is under basic investigation.


Sujet(s)
Syndrome de Guillain-Barré , Humains , Femelle , Syndrome de Guillain-Barré/diagnostic , Syndrome de Guillain-Barré/traitement médicamenteux , Jeune adulte , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B/complications , Dengue/diagnostic , Dengue/complications , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/complications , Leucémie-lymphome lymphoblastique à précurseurs B et T/diagnostic
10.
Invest New Drugs ; 42(3): 299-308, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38662275

RÉSUMÉ

Blinatumomab is efficacious in patients with B-cell acute lymphoblastic leukemia (B-ALL), yet limited real-world data exists in this context. This retrospective study provided real-world data on the treatment pattern, effectiveness, and safety of blinatumomab in Chinese patients with newly diagnosed (ND) and relapsed/refractory (R/R) B-ALL. Patients with B-ALL who received at least one dose of blinatumomab in frontline or R/R settings between August 2021 and June 2023 were included. The primary outcome was the treatment pattern of blinatumomab. Key secondary outcomes included complete remission (CR)/CR with incomplete blood cell recovery (CRi) rate, minimal residual disease (MRD) negativity, median event-free survival (EFS), and safety. The study included 96 patients with B-ALL; 53 (55.2%) patients were in the ND group and 43 (44.8%) patients were in the R/R group. The median treatment duration was one cycle (range: 1-5). Most patients underwent chemotherapies, allo-HSCT, or experimental CAR-T following blinatumomab. The ND patients using blinatumomab induction therapy achieved 100% CR/CRi rate; 87.2% achieved MRD negativity within two cycles of blinatumomab. In R/R re-induction patients, the CR/CRi rate was 50%; MRD negativity rate was 64.2%. In R/R patients using blinatumomab for consolidation, MRD negativity rate was 90.9%. The median EFS was not reached in both ND and R/R patients; 1-year EFS rate was 90.8% (95% CI: 67%, 97%) and 55.1% (95% CI: 30%, 74%), respectively. Grade ≥ 3 adverse events were observed in 12.5% patients. Blinatumomab was found to be effective with a tolerable safety profile in real world setting.


Sujet(s)
Anticorps bispécifiques , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Anticorps bispécifiques/effets indésirables , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/administration et posologie , Mâle , Femelle , Adulte , Adulte d'âge moyen , Études rétrospectives , Adolescent , Jeune adulte , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Sujet âgé , Enfant , Antinéoplasiques/usage thérapeutique , Antinéoplasiques/effets indésirables , Antinéoplasiques/administration et posologie , Chine , Résultat thérapeutique , Maladie résiduelle , Enfant d'âge préscolaire , Induction de rémission , Peuples d'Asie de l'Est
11.
Cancer Med ; 13(8): e7172, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38651186

RÉSUMÉ

BACKGROUND: Quantitative measurement of minimal residual disease (MRD) is the "gold standard" for estimating the response to therapy in childhood B-cell precursor acute lymphoblastic leukemia (BCP-ALL). Nevertheless, the speed of the MRD response differs for different cytogenetic subgroups. Here we present results of MRD measurement in children with BCP-ALL, in terms of genetic subgroups with relation to clinically defined risk groups. METHODS: A total of 485 children with non-high-risk BCP-ALL with available cytogenetic data and MRD studied at the end-of-induction (EOI) by multicolor flow cytometry (MFC) were included. All patients were treated with standard-risk (SR) of intermediate-risk (ImR) regimens of "ALL-MB 2008" reduced-intensity protocol. RESULTS AND DISCUSSION: Among all study group patients, 203 were found to have low-risk cytogenetics (ETV6::RUNX1 or high hyperdiploidy), while remaining 282 children were classified in intermediate cytogenetic risk group. For the patients with favorable and intermediate risk cytogenetics, the most significant thresholds for MFC-MRD values were different: 0.03% and 0.04% respectively. Nevertheless, the most meaningful thresholds were different for clinically defined SR and ImR groups. For the SR group, irrespective to presence/absence of favorable genetic lesions, MFC-MRD threshold of 0.1% was the most clinically valuable, although for ImR group the most informative thresholds were different in patients from low-(0.03%) and intermediate (0.01%) cytogenetic risk groups. CONCLUSION: Our data show that combining clinical risk factors with MFC-MRD measurement is the most useful tool for risk group stratification of children with BCP-ALL in the reduced-intensity protocols. However, this algorithm can be supplemented with cytogenetic data for part of the ImR group.


Sujet(s)
Cytométrie en flux , Maladie résiduelle , Humains , Maladie résiduelle/génétique , Enfant , Cytométrie en flux/méthodes , Mâle , Femelle , Enfant d'âge préscolaire , Adolescent , Nourrisson , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Analyse cytogénétique/méthodes , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/diagnostic , Sous-unité alpha 2 du facteur CBF/génétique
12.
Leuk Res ; 141: 107506, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38663165

RÉSUMÉ

Significant heterogeneity has been reported in outcome of Acute lymphoblastic leukemia with t(1;19)(q23;p13)/TCF3::PBX1 in adolescents and adults leading to a lack of consensus on precise risk stratification. We evaluated clinical outcome of 17 adult ALL cases (≥15 years) with this genotype treated on intensive regimes.13/17 received COG0232 and 4/17 cases received UK-ALL protocol. All achieved CR (100%) with above treatment. End of induction MRD was evaluated in 14/17 cases of which 11 (78.5%) achieved MRD negativity. Total nine patients relapsed (7 marrows, 2 CNS). Overall survival at 2 years was 53.3%. The 2 year estimated PFS was 42.9%. The 2 years CIR was 54.2%. Adults with this genotype perform poorly despite early favorable response. Incorporation of novel immunotherapies and prompt HSCT should be strongly considered with this genotype. Targeted NGS panels for additional genetic aberrations can further help in risk stratifying and guiding therapy for this genotype.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Humains , Mâle , Adulte , Femelle , Adolescent , Adulte d'âge moyen , Jeune adulte , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/mortalité , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Protéines de fusion oncogènes/génétique , Translocation génétique , Chromosomes humains de la paire 19/génétique , Taux de survie , Pronostic , Résultat thérapeutique
13.
Hematology ; 29(1): 2344998, 2024 Dec.
Article de Anglais | MEDLINE | ID: mdl-38666535

RÉSUMÉ

OBJECTIVES: Relapsed/refractory acute B-cell lymphoblastic leukemia (R/R B-ALL) often responds poorly to induction chemotherapy. However, recent research has shown a novel and effective drug treatment for R/R B-ALL. METHODS: A total of eight patients with R/R B-ALL were enrolled in the study from November 2021 to August 2022. All patients received chemotherapy based on a combination regimen of venetoclax and azacitidine. The regimen was as follows venetoclax 100 mg d1, 200 mg d2, 400 mg d3-14, azacitidine 75 mg/m2 d1-7. RESULTS: Five of eight patients achieved very deep and complete remission (CR) with minimal residual disease (MRD) less than 0.1%. One patient achieved partial remission. Two patients did not achieve remission. There were no serious adverse events and all patients were well tolerated. Three patients were eligible for consolidation chemotherapy and were bridged to CAR-T therapy. CONCLUSIONS: The combined regimen of venetoclax and azacitidine may be beneficial for patients with R/R B-ALL.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique , Azacitidine , Composés hétérocycliques bicycliques , Sulfonamides , Humains , Azacitidine/usage thérapeutique , Azacitidine/administration et posologie , Composés hétérocycliques bicycliques/usage thérapeutique , Composés hétérocycliques bicycliques/administration et posologie , Femelle , Mâle , Sulfonamides/usage thérapeutique , Sulfonamides/administration et posologie , Adulte d'âge moyen , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Adulte , Sujet âgé , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux
14.
Cancer Cell ; 42(4): 552-567.e6, 2024 Apr 08.
Article de Anglais | MEDLINE | ID: mdl-38593781

RÉSUMÉ

Leukemia can arise at various stages of the hematopoietic differentiation hierarchy, but the impact of developmental arrest on drug sensitivity is unclear. Applying network-based analyses to single-cell transcriptomes of human B cells, we define genome-wide signaling circuitry for each B cell differentiation stage. Using this reference, we comprehensively map the developmental states of B cell acute lymphoblastic leukemia (B-ALL), revealing its strong correlation with sensitivity to asparaginase, a commonly used chemotherapeutic agent. Single-cell multi-omics analyses of primary B-ALL blasts reveal marked intra-leukemia heterogeneity in asparaginase response: resistance is linked to pre-pro-B-like cells, with sensitivity associated with the pro-B-like population. By targeting BCL2, a driver within the pre-pro-B-like cell signaling network, we find that venetoclax significantly potentiates asparaginase efficacy in vitro and in vivo. These findings demonstrate a single-cell systems pharmacology framework to predict effective combination therapies based on intra-leukemia heterogeneity in developmental state, with potentially broad applications beyond B-ALL.


Sujet(s)
Leucémies , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Asparaginase/pharmacologie , Pharmacologie des réseaux , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Transduction du signal , Leucémies/traitement médicamenteux
15.
Br J Haematol ; 204(4): 1367-1374, 2024 Apr.
Article de Anglais | MEDLINE | ID: mdl-38444113

RÉSUMÉ

Literature regarding prognostic relevance of CD20 antigen expression among paediatric B-lineage acute lymphoblastic leukaemia (B-ALL) patients is sparse and contradictory. We analysed clinical laboratory parameters and survival characteristics pertinent to CD20 expression among 224 treatment-naïve paediatric B-ALL patients. 50% patients had CD20 expression (CD20+ B-ALL). There was no difference in the clinical & laboratory presentation and end of induction measurable residual disease (EOI-MRD) status according to CD20 expression. As compared to CD20- B-ALL patients, CD20+ B-ALL patients had two times more relapse (16% vs. 29%, p = 0.034), inferior relapse-free survival (79% vs. 66%, p = 0.025) but no difference in overall survival (75% vs. 69%, p = 0.126). Similar to high-risk NCI status and EOI-MRD positivity, CD20 expression was an independent predictor for inferior relapse-free survival (HR: 1.860, 95% CI: 1.008-3.432, p = 0.047). Compared to baseline, there was a significant increase in CD20-expressing EOI-residual blasts among CD20- B-ALL patients (5% vs. 13%, p = 0.001). EOI residual blasts of both CD20+ and CD20- patients had three times increased normalized CD20 expression intensity (nCD20), with the intensity among CD20- B-ALL patients reaching the pretreatment nCD20 of CD20+ B-ALL patients (4.9 vs. 3.6, p = 0.666). Rituximab can be considered in managing EOI-MRD-positive CD20- B-ALL patients as the residual blasts of these patients have quantitative and qualitative increases in CD20 expression.


Sujet(s)
Lymphome B , Leucémie-lymphome lymphoblastique à précurseurs B , Leucémie-lymphome lymphoblastique à précurseurs B et T , Enfant , Humains , Antigènes CD20 , Récidive tumorale locale , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Rituximab/usage thérapeutique , Maladie résiduelle
16.
Chemotherapy ; 69(2): 104-107, 2024.
Article de Anglais | MEDLINE | ID: mdl-38508148

RÉSUMÉ

INTRODUCTION: With the increasing use of blinatumomab in relapsed or refractory B-cell precursor acute lymphoblastic leukemia (ALL), including minimal residual disease (MRD)-positive cases, awareness of its adverse effects has gradually improved. Pneumocystis jirovecii pneumonia (PCP) associated with blinatumomab therapy is rare. CASE PRESENTATION: We present a case of PCP in a patient undergoing blinatumomab therapy. A 70-year-old female diagnosed with Philadelphia-like CRLF2 overexpression B-cell precursor ALL received blinatumomab as consolidation therapy after achieving complete remission with prior induction chemotherapy. On the second day of blinatumomab infusion, she developed intermittent low-grade fever, and chest computed tomography (CT) revealed subtle infiltrates and nodules. Despite empiric trimethoprim-sulfamethoxazole (TMP-SMX) prophylaxis, she progressed to significant shortness of breath and type I respiratory failure, with increased lactate dehydrogenase and ß-D-glucan assays. Chest CT showed diffuse ground-glass opacities with scattered small nodules. The dry cough prompted next-generation sequencing of peripheral blood, which tested positive for pneumocystis jirovecii without evidence of other pathogens. Consequently, the patient was diagnosed with PCP. The first cycle of blinatumomab had to be discontinued, and therapeutic dosages of TMP-SMX and dexamethasone were administered, resulting in full recovery and stable condition during follow-ups. CONCLUSION: PCP is rare in B-cell precursor ALL patients receiving blinatumomab therapy but manifests with early onset and rapid disease progression. Despite prophylaxis, PCP infection cannot be ignored during blinatumomab therapy. Therefore, heightened attention is warranted when using blinatumomab therapy.


Sujet(s)
Anticorps bispécifiques , Pneumocystis carinii , Pneumonie à Pneumocystis , Humains , Femelle , Pneumonie à Pneumocystis/traitement médicamenteux , Pneumonie à Pneumocystis/diagnostic , Sujet âgé , Anticorps bispécifiques/usage thérapeutique , Anticorps bispécifiques/effets indésirables , Pneumocystis carinii/isolement et purification , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/complications , Tomodensitométrie , Association triméthoprime-sulfaméthoxazole/usage thérapeutique , Association triméthoprime-sulfaméthoxazole/effets indésirables
18.
Transplant Cell Ther ; 30(5): 520.e1-520.e12, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38462215

RÉSUMÉ

BACKGROUND: Blinatumomab, a bispecific monoclonal antibody, effectively controls refractory B cell acute lymphoblastic leukemia (ALL) and promotes measurable residual disease (MRD) negativity. This study investigated the impact of pretransplant blinatumomab on allogeneic hematopoietic cell transplantation (HCT) outcomes in B cell ALL patients. METHODS: We analyzed the effect of pretransplant blinatumomab on transplant outcomes of 117 adults undergoing allogeneic HCT for B cell ALL at Princess Margaret Hospital, Toronto, between 2010 and 2021. Outcomes assessed included overall survival (OS), graft-versus-host disease and relapse-free survival (GRFS), cumulative incidences of relapse (CIR), and nonrelapse mortality (NRM). RESULTS: The median follow-up was 36 months. Thirty-one participants (26.5%) received blinatumomab. Blinatumomab group had higher proportions of individuals with high disease risk index, primary induction failure and was more likely to receive dual T cell depletion with antithymocyte globulin and post-transplant cyclophosphamide. Two-year OS, GRFS, NRM, and CIR in the blinatumomab and nonblinatumomab groups were, respectively: 65.4% versus 45.6% (P = .05), 42.2% versus 17.3% (P = .01), 3.2% versus 43.0% (P = .007) and 34.4% versus 14.4% (P = .02). Blinatumomab was associated with a lower incidence of day-100 grade 2 to 4 and grade 3 to 4 acute graft-versus-host disease (aGVHD): 27.5% versus 56.7% (P = .009), and 10.9% versus 34.7% (P = .04), respectively. Multivariate analysis confirmed the association between pretransplant blinatumomab and improved OS and NRM. CONCLUSIONS: Pretransplant blinatumomab is associated with improved OS and lower risk of NRM in B cell ALL patients undergoing allogeneic HCT, likely reflecting lower burden of treatment-related toxicity in this population. Larger prospective trials are warranted to validate our findings.


Sujet(s)
Anticorps bispécifiques , Transplantation de cellules souches hématopoïétiques , Humains , Anticorps bispécifiques/usage thérapeutique , Mâle , Femelle , Adulte , Adulte d'âge moyen , Transplantation homologue , Jeune adulte , Résultat thérapeutique , Adolescent , Sujet âgé , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B/mortalité , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Maladie du greffon contre l'hôte , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie
19.
Cancer Med ; 13(5): e7062, 2024 Mar.
Article de Anglais | MEDLINE | ID: mdl-38491815

RÉSUMÉ

BACKGROUND: Blinatumomab early-line treatment in B-cell precursor acute lymphoblastic leukemia (B-ALL) might improve clinical outcomes. METHODS: We conducted a retrospective real-world cohort analysis in 20 newly diagnosed B-ALL patients who received reduced-dose chemotherapy (idarubicin, vindesine, and dexamethasone) for 1-3 weeks, followed by blinatumomab for 1-4 weeks as an induction therapy. RESULTS: At the end of the induction therapy, a complete remission rate of 100% was achieved; 17 (85%) patients were minimal residual disease (MRD) negative (<1 × 10-4 ). Adverse events (AEs) were reported in 12 (60%) patients-43.8% were grade 1-2 and 56.2% were grade 3-4. No incidence of neurotoxicity or grade ≥3 cytokine release syndrome was reported. CONCLUSIONS: Blinatumomab demonstrated a significant improvement in clinical outcomes in patients with newly diagnosed B-ALL irrespective of their poor-risk factor status and the pretreatment blast burden.


Sujet(s)
Anticorps bispécifiques , Lymphome de Burkitt , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Études rétrospectives , Chimiothérapie d'induction , Anticorps bispécifiques/effets indésirables , Leucémie-lymphome lymphoblastique à précurseurs B/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Lymphome de Burkitt/traitement médicamenteux
20.
Cell Death Dis ; 15(3): 216, 2024 Mar 14.
Article de Anglais | MEDLINE | ID: mdl-38485947

RÉSUMÉ

Despite progressive improvements in the survival rate of pediatric B-cell lineage acute lymphoblastic leukemia (B-ALL), chemoresistance-induced disease progression and recurrence still occur with poor prognosis, thus highlighting the urgent need to eradicate drug resistance in B-ALL. The 6-mercaptopurine (6-MP) is the backbone of ALL combination chemotherapy, and resistance to it is crucially related to relapse. The present study couples chemoresistance in pediatric B-ALL with histidine metabolism deficiency. Evidence was provided that histidine supplementation significantly shifts the 6-MP dose-response in 6-MP-resistant B-ALL. It is revealed that increased tetrahydrofolate consumption via histidine catabolism partially explains the re-sensitization ability of histidine. More importantly, this work provides fresh insights into that desuccinylation mediated by SIRT5 is an indispensable and synergistic requirement for histidine combination therapy against 6-MP resistance, which is undisclosed previously and demonstrates a rational strategy to ameliorate chemoresistance and protect pediatric patients with B-ALL from disease progression or relapse.


Sujet(s)
Lymphome de Burkitt , Leucémie-lymphome lymphoblastique à précurseurs B , Leucémie-lymphome lymphoblastique à précurseurs B et T , Sirtuines , Humains , Enfant , Mercaptopurine/pharmacologie , Mercaptopurine/usage thérapeutique , Histidine/usage thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B et T/traitement médicamenteux , Leucémie-lymphome lymphoblastique à précurseurs B/traitement médicamenteux , Lymphome de Burkitt/traitement médicamenteux , Récidive , Évolution de la maladie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...