Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 3.287
Filtrer
1.
Zhonghua Xue Ye Xue Za Zhi ; 45(4): 339-344, 2024 Apr 14.
Article de Chinois | MEDLINE | ID: mdl-38951060

RÉSUMÉ

Objective: Exploring the efficacy and safety of bridging blinatumomab (BiTE) in combination with chimeric antigen receptor T (CAR-T) cell therapy for the treatment of adult patients with acute B-cell lymphoblastic leukemia (B-ALL) . Methods: Clinical data from 36 adult B-ALL patients treated at the First Affiliated Hospital of Suzhou University from August 2018 to May 2023 were retrospectively analyzed. A total of 36 cases were included: 18 men and 18 women. The median age was 43.5 years (21-72 years). Moreover, 21 cases of Philadelphia chromosome-positive acute lymphoblastic leukemia were reported, and 16 of these cases were relapsed or refractory. Eighteen patients underwent blinatumomab bridging followed by CAR-T cell therapy, and 18 patients received CAR-T cell therapy. This study analyzed the efficacy and safety of treatment in two groups of patients. Results: In the BiTE bridge-to-CAR-T group, 16 patients achieved complete remission (CR) after BiTE immunotherapy, with a CR rate of 88.9%. One month after bridging CAR-T therapy, bone marrow examination showed a CR rate of 100.0%, and the minimal residual disease (MRD) negativity rate was higher than the nonbridging therapy group (94.4% vs. 61.1%, Fisher, P=0.041). The incidence of cytokine release syndrome and other adverse reactions in the BiTE bridge-to-CAR-T group was lower than that in the nonbridging therapy group (11.1% vs. 50.0%, Fisher, P=0.027). The follow-up reveals that 13 patients continued to maintain MRD negativity, and five patients experienced relapse 8.40 months (2.57-10.20 months) after treatment. Two of five patients with relapse achieved CR after receiving the second CAR-T cell therapy. In the nonbridging therapy group, 10 patients maintained continuous MRD negativity, 7 experienced relapse, and 6 died. The 1 year overall survival rate in the BiTE bridge-to-CAR-T group was higher than that in the nonbridging therapy group, with a statistically significant difference at the 0.1 level (88.9%±10.5% vs. 66.7%±10.9%, P=0.091) . Conclusion: BiTE bridging CAR-T cell therapy demonstrates excellent efficacy in adult B-ALL treatment, with a low recent recurrence rate and ongoing assessment of long-term efficacy during follow-up.


Sujet(s)
Anticorps bispécifiques , Immunothérapie adoptive , Humains , Mâle , Adulte , Femelle , Anticorps bispécifiques/administration et posologie , Adulte d'âge moyen , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Études rétrospectives , Jeune adulte , Sujet âgé , Résultat thérapeutique , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B et T/thérapie
2.
J Exp Clin Cancer Res ; 43(1): 179, 2024 Jun 27.
Article de Anglais | MEDLINE | ID: mdl-38926853

RÉSUMÉ

BACKGROUND: Enhancer reprogramming plays a significant role in the heterogeneity of cancer. However, we have limited knowledge about the impact of chromatin remodeling in B-Cell Precursor Acute Lymphoblastic Leukemia (BCP-ALL) patients, and how it affects tumorigenesis and drug response. Our research focuses on investigating the role of enhancers in sustaining oncogenic transformation in children with BCP-ALL. METHODS: We used ATAC-seq to study the accessibility of chromatin in pediatric BCP-ALL at three different stages-onset, remission, and relapse. Using a combination of computational and experimental methods, we were able to analyze the accessibility landscape and focus on the most significant cis-regulatory sites. These sites were then functionally validated through the use of Promoter capture Hi-C in a primary cell line model called LAL-B, followed by RNA-seq and genomic deletion of target sites using CRISPR-Cas9 editing. RESULTS: We found that enhancer activity changes during cancer progression and is mediated by the production of enhancer RNAs (eRNAs). CRISPR-Cas9-mediated validation of previously unknown eRNA productive enhancers demonstrated their capability to control the oncogenic activities of the MYB and DCTD genes. CONCLUSIONS: Our findings directly support the notion that productive enhancer engagement is a crucial determinant of the BCP-ALL and highlight the potential of enhancers as therapeutic targets in pediatric BCP-ALL.


Sujet(s)
Transformation cellulaire néoplasique , Évolution de la maladie , Éléments activateurs (génétique) , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B/métabolisme , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Enfant
5.
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
6.
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
7.
Haematologica ; 109(6): 1689-1699, 2024 Jun 01.
Article de Anglais | MEDLINE | ID: mdl-38832424

RÉSUMÉ

Chimeric antigen receptor (CAR) T-cell therapy has emerged as a breakthrough cancer therapy over the past decade. Remarkable outcomes in B-cell lymphoproliferative disorders and multiple myeloma have been reported in both pivotal trials and real-word studies. Traditionally, the use of a patient's own (autologous) T cells to manufacture CAR products has been the standard practice. Nevertheless, this approach has some drawbacks, including manufacturing delays, dependence on the functional fitness of the patient's T cells, which can be compromised by both the disease and prior therapies, and contamination of the product with blasts. A promising alternative is offered by the development of allogeneic CAR-cell products. This approach has the potential to yield more efficient drug products and enables the use of effector cells with negligible alloreactive potential and a significant CAR-independent antitumor activity through their innate receptors (i.e., natural killer cells, γδ T cells and cytokine induced killer cells). In addition, recent advances in genome editing tools offer the potential to overcome the primary challenges associated with allogeneic CAR T-cell products, namely graft-versus-host disease and host allo-rejection, generating universal, off-the-shelf products. In this review, we summarize the current pre-clinical and clinical approaches based on allogeneic CAR T cells, as well as on alternative effector cells, which represent exciting opportunities for multivalent approaches and optimized antitumor activity.


Sujet(s)
Immunothérapie adoptive , Leucémie-lymphome lymphoblastique à précurseurs B , Récepteurs chimériques pour l'antigène , Humains , Récepteurs chimériques pour l'antigène/immunologie , Récepteurs chimériques pour l'antigène/génétique , Immunothérapie adoptive/méthodes , Immunothérapie adoptive/effets indésirables , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Enfant , Transplantation homologue , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Résultat thérapeutique
8.
J Cancer Res Clin Oncol ; 150(6): 314, 2024 Jun 22.
Article de Anglais | MEDLINE | ID: mdl-38907739

RÉSUMÉ

PURPOSE: Leukemia-associated fusion genes are closely related to the occurrence, development, diagnosis, and treatment of leukemia. DNA microarrays and second-generation sequencing have discovered multiple B-ALL fusion genes. We identified a novel MEF2C::SS18L1 fusion gene in a child diagnosed with B-ALL. This study investigates the oncogenicity and prognosis of this fusion gene in B-ALL. METHODS: A child with B-ALL who has a MEF2C::SS18L1 fusion is reported as a newly discovered case. Compared the breakpoints, structural domains, clinical phenotypes, and differential expression genes of MEF2C::SS18L1 and MEF2D::SS18.Using "ONCOFUSE" software, the carcinogenicity of MEF2C::SS18L1 is predicted. Using whole transcriptome sequencing, we analyze the breakpoints and the secondary structure of the fusion protein. Further, we compared the structures, differentially expressed genes, and clinical phenotypes of MEF2D and MEF2C fusion genes by DESeq, GO functional enrichment, and flow cytometry immunophenotyping analysis. RESULTS: Whole transcriptome sequencing identified a MEF2C::SS18L1 fusion transcript in a 3-year-old child with B-ALL. The MADS box, MEF structural domain, HJURP_C structural domain, and TAD I structural domain of MEF2C, and the QPGY structural domain of SS18L1, make up the fusion protein. "Oncofuse" found a 0.99 Bayesian probability that the fusion gene drives cancer. The breakpoint positions, fusion protein secondary structures, differentially expressed genes, and clinical characteristics of this patient were identical to those with MEF2D::SS18 fusion gene. CONCLUSION: We identified a novel MEF2C::SS18L1 fusion gene in childhood ALL, which shares similar structural and clinical characteristics with MEF2D::SS18. Further studies with more samples should be conducted in future.


Sujet(s)
Facteurs de transcription MEF2 , Protéines de fusion oncogènes , Humains , Facteurs de transcription MEF2/génétique , Protéines de fusion oncogènes/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Mâle , Enfant d'âge préscolaire , Enfant , Femelle
9.
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
10.
Mol Biol Rep ; 51(1): 749, 2024 Jun 14.
Article de Anglais | MEDLINE | ID: mdl-38874800

RÉSUMÉ

Background The incidence of various types of cancers, including leukemia, is on the rise and many challenges in both drug resistance and complications related to chemotherapy appeared. Recently, the development and application of extracellular vesicles (EV) such as exosomes in the management of cancers, especially leukemia, holds great significance. In this article, we extracted exosomes from NALM6 cells and assessed their regulatory effects on proliferation and apoptosis in mesenchymal stem cells (MSCs). Method and result We first verified the exosomes using various techniques, including flow cytometry, transient electron microscopy, dynamic light scattering (DLS), and BCA protein assay. Then MTT analysis and flowcytometry (apoptosis and cell cycle assay) besides gene expressions were employed to determine the state of MSC proliferations. The results indicated that exosome-specific pan markers like CD9, CD63, and CD81 were present. Through DLS, we found out that the mean size of the exosomes was 89.68 nm. The protein content was determined to be 956.292 µg/ml. Analysis of MTT, flow cytometry (cell cycle and apoptosis assay), and RT-qPCR showed that in the dose of 50 µg/ml the proliferation of MSCs was increased significantly (p-value < 0.05). Conclusion All these data showed that exosomes use several signaling pathways to increase the MSCs' proliferation and drug resistance, ultimately leading to high mortalities and morbidities of acute lymphoblastic leukemia.


Sujet(s)
Apoptose , Prolifération cellulaire , Exosomes , Cellules souches mésenchymateuses , Exosomes/métabolisme , Cellules souches mésenchymateuses/métabolisme , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Humains , Prolifération cellulaire/effets des médicaments et des substances chimiques , Apoptose/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Antigène CD9/métabolisme , Antigène CD9/génétique , Cycle cellulaire/effets des médicaments et des substances chimiques , Cycle cellulaire/génétique , Leucémie-lymphome lymphoblastique à précurseurs B et T/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B et T/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B et T/génétique , Antigène CD63/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/métabolisme , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Leucémie-lymphome lymphoblastique à précurseurs B/génétique
11.
Cancer Lett ; 596: 217018, 2024 Aug 01.
Article de Anglais | MEDLINE | ID: mdl-38844062

RÉSUMÉ

Relapse and treatment resistance pose significant challenges in the management of pediatric B cell acute lymphoblastic leukemia (B-ALL) and acute myeloid leukemia (AML). The efficacy of immunotherapy in leukemia remains limited due to factors such as the immunosuppressive tumor microenvironment (TME) and lack of suitable immunotherapeutic targets. Thus, an in-depth characterization of the TME in pediatric leukemia is warranted to improve the efficacy of immunotherapy. Here, we used single-cell RNA sequencing (scRNA-seq) to characterize the TME of pediatric B-ALL and AML, focusing specifically on bone-marrow-derived T cells. Moreover, we investigated the transcriptome changes during the initiation, remission, and relapse stages of pediatric AML. Our findings revealed that specific functional expression programs correlated with fluctuations in various T cell subsets, which may be associated with AML progression and relapse. Furthermore, our analysis of cellular communication networks led to the identification of VISTA, CD244, and TIM3 as potential immunotherapeutic targets in pediatric AML. Finally, we detected elevated proportions of γδ T cells and associated functional genes in samples from pediatric patients diagnosed with B-ALL and AML, which could inform the development of novel therapeutic approaches, potentially focusing on γδ T cells.


Sujet(s)
Leucémie aigüe myéloïde , Analyse sur cellule unique , Microenvironnement tumoral , Humains , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/génétique , Analyse sur cellule unique/méthodes , Enfant , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/immunologie , Leucémie aigüe myéloïde/anatomopathologie , Transcriptome , Récepteur cellulaire-2 du virus de l'hépatite A/génétique , Récepteur cellulaire-2 du virus de l'hépatite A/métabolisme , Analyse de profil d'expression de gènes/méthodes , Enfant d'âge préscolaire , Mâle , Femelle , Antigènes B7/génétique , Adolescent , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/immunologie , Leucémie-lymphome lymphoblastique à précurseurs B/anatomopathologie , Régulation de l'expression des gènes dans la leucémie
12.
Int J Mol Sci ; 25(12)2024 Jun 15.
Article de Anglais | MEDLINE | ID: mdl-38928295

RÉSUMÉ

The genomic analyses of pediatric acute lymphoblastic leukemia (ALL) subtypes, particularly T-cell and B-cell lineages, have been pivotal in identifying potential therapeutic targets. Typical genomic analyses have directed attention toward the most commonly mutated genes. However, assessing the contribution of mutations to cancer phenotypes is crucial. Therefore, we estimated the cancer effects (scaled selection coefficients) for somatic substitutions in T-cell and B-cell cohorts, revealing key insights into mutation contributions. Cancer effects for well-known, frequently mutated genes like NRAS and KRAS in B-ALL were high, which underscores their importance as therapeutic targets. However, less frequently mutated genes IL7R, XBP1, and TOX also demonstrated high cancer effects, suggesting pivotal roles in the development of leukemia when present. In T-ALL, KRAS and NRAS are less frequently mutated than in B-ALL. However, their cancer effects when present are high in both subtypes. Mutations in PIK3R1 and RPL10 were not at high prevalence, yet exhibited some of the highest cancer effects in individual T-cell ALL patients. Even CDKN2A, with a low prevalence and relatively modest cancer effect, is potentially highly relevant for the epistatic effects that its mutated form exerts on other mutations. Prioritizing investigation into these moderately frequent but potentially high-impact targets not only presents novel personalized therapeutic opportunities but also enhances the understanding of disease mechanisms and advances precision therapeutics for pediatric ALL.


Sujet(s)
Mutation , Humains , Enfant , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/épidémiologie , Leucémie-lymphome lymphoblastique à précurseurs T/génétique , Lymphocytes T/immunologie , Lymphocytes T/métabolisme , Lymphocytes B/immunologie , Lymphocytes B/métabolisme
14.
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
15.
Ann Hematol ; 103(7): 2551-2556, 2024 Jul.
Article de Anglais | MEDLINE | ID: mdl-38724656

RÉSUMÉ

Chimeric antigen receptor T (CAR-T) cells therapy is a milestone achievement in the immunotherapy of relapsed and refractory (R/R) B cell acute lymphoblastic leukemia (B-ALL). However, some patients treated with CAR-T cells do not achieve complete remission, the mechanisms of which have not been elucidated. In the present study, we report a 9-year-old pediatric patient with refractory B-ALL received a triple infusion of autologous CD19 CAR-T cells therapy after the second relapse. CAR-T cells expanded in the peripheral blood and bone marrow. However, the patient did not achieve complete remission, indicating a lack of response to CAR-T cells therapy. Analysis of etiological factors revealed that the number of CD4 and CD8 double-negative T (DNT) cells was significantly upregulated in the peripheral blood, bone marrow, and autologous CAR-T cells products. In conclusiont, these findings indicate that DNT cells mediated resistance to CAR-T cells therapy in this pediatric patient with R/R B-ALL.


Sujet(s)
Immunothérapie adoptive , Leucémie-lymphome lymphoblastique à précurseurs B , Humains , Enfant , Immunothérapie adoptive/méthodes , Leucémie-lymphome lymphoblastique à précurseurs B/thérapie , Antigènes CD19/immunologie , Récepteurs chimériques pour l'antigène/immunologie , Mâle , Récidive , Résistance aux médicaments antinéoplasiques , Femelle
16.
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
17.
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
18.
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
20.
EBioMedicine ; 104: 105171, 2024 Jun.
Article de Anglais | MEDLINE | ID: mdl-38810562

RÉSUMÉ

BACKGROUND: The increasing volume and intricacy of sequencing data, along with other clinical and diagnostic data, like drug responses and measurable residual disease, creates challenges for efficient clinical comprehension and interpretation. Using paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) as a use case, we present an artificial intelligence (AI)-assisted clinical framework clinALL that integrates genomic and clinical data into a user-friendly interface to support routine diagnostics and reveal translational insights for hematologic neoplasia. METHODS: We performed targeted RNA sequencing in 1365 cases with haematological neoplasms, primarily paediatric B-cell precursor acute lymphoblastic leukaemia (BCP-ALL) from the AIEOP-BFM ALL study. We carried out fluorescence in situ hybridization (FISH), karyotyping and arrayCGH as part of the routine diagnostics. The analysis results of these assays as well as additional clinical information were integrated into an interactive web interface using Bokeh, where the main graph is based on Uniform Manifold Approximation and Projection (UMAP) analysis of the gene expression data. At the backend of the clinALL, we built both shallow machine learning models and a deep neural network using Scikit-learn and PyTorch respectively. FINDINGS: By applying clinALL, 78% of undetermined patients under the current diagnostic protocol were stratified, and ambiguous cases were investigated. Translational insights were discovered, including IKZF1plus status dependent subpopulations of BCR::ABL1 positive patients, and a subpopulation within ETV6::RUNX1 positive patients that has a high relapse frequency. Our best machine learning models, LDA and PASNET-like neural network models, achieve F1 scores above 97% in predicting patients' subgroups. INTERPRETATION: An AI-assisted clinical framework that integrates both genomic and clinical data can take full advantage of the available data, improve point-of-care decision-making and reveal clinically relevant insights promptly. Such a lightweight and easily transferable framework works for both whole transcriptome data as well as the cost-effective targeted RNA-seq, enabling efficient and equitable delivery of personalized medicine in small clinics in developing countries. FUNDING: German Ministry of Education and Research (BMBF), German Research Foundation (DFG) and Foundation for Polish Science.


Sujet(s)
Intelligence artificielle , , Humains , Tumeurs hématologiques/génétique , Tumeurs hématologiques/diagnostic , Leucémie-lymphome lymphoblastique à précurseurs B/génétique , Leucémie-lymphome lymphoblastique à précurseurs B/diagnostic , Biologie informatique/méthodes , Enfant , Hybridation fluorescente in situ/méthodes , Femelle , Mâle , Marqueurs biologiques tumoraux/génétique , Analyse de profil d'expression de gènes/méthodes
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...