Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 6.251
Filtrer
1.
J Clin Invest ; 134(19)2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39352393

RÉSUMÉ

Clonal hematopoiesis (CH), the expansion of hematopoietic stem cells and their progeny driven by somatic mutations in leukemia-associated genes, is a common phenomenon that rises in prevalence with advancing age to affect most people older than 70 years. CH remains subclinical in most carriers, but, in a minority, it progresses to a myeloid neoplasm, such as acute myeloid leukemia, myelodysplastic syndrome, or myeloproliferative neoplasm. Over the last decade, advances in our understanding of CH, its molecular landscape, and the risks associated with different driver gene mutations have culminated in recent developments that allow for a more precise estimation of myeloid neoplasia risk in CH carriers. In turn, this is leading to the development of translational and clinical programs to intercept and prevent CH from developing into myeloid neoplasia. Here, we give an overview of the spectrum of CH driver mutations, what is known about their pathophysiology, and how this informs the risk of incident myeloid malignancy.


Sujet(s)
Hématopoïèse clonale , Tumeurs hématologiques , Mutation , Humains , Hématopoïèse clonale/génétique , Tumeurs hématologiques/génétique , Tumeurs hématologiques/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/anatomopathologie , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/anatomopathologie , Syndromes myéloprolifératifs/métabolisme , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/anatomopathologie
3.
N Engl J Med ; 391(14): 1287-1301, 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39383458

RÉSUMÉ

BACKGROUND: Gene therapy with elivaldogene autotemcel (eli-cel) consisting of autologous CD34+ cells transduced with lentiviral vector containing ABCD1 complementary DNA (Lenti-D) has shown efficacy in clinical studies for the treatment of cerebral adrenoleukodystrophy. However, the risk of oncogenesis with eli-cel is unclear. METHODS: We performed integration-site analysis, genetic studies, flow cytometry, and morphologic studies in peripheral-blood and bone marrow samples from patients who received eli-cel therapy in two completed phase 2-3 studies (ALD-102 and ALD-104) and an ongoing follow-up study (LTF-304) involving the patients in both ALD-102 and ALD-104. RESULTS: Hematologic cancer developed in 7 of 67 patients after the receipt of eli-cel (1 of 32 patients in the ALD-102 study and 6 of 35 patients in the ALD-104 study): myelodysplastic syndrome (MDS) with unilineage dysplasia in 2 patients at 14 and 26 months; MDS with excess blasts in 3 patients at 28, 42, and 92 months; MDS in 1 patient at 36 months; and acute myeloid leukemia (AML) in 1 patient at 57 months. In the 6 patients with available data, predominant clones contained lentiviral vector insertions at multiple loci, including at either MECOM-EVI1 (MDS and EVI1 complex protein EVI1 [ecotropic virus integration site 1], in 5 patients) or PRDM16 (positive regulatory domain zinc finger protein 16, in 1 patient). Several patients had cytopenias, and most had vector insertions in multiple genes within the same clone; 6 of the 7 patients also had somatic mutations (KRAS, NRAS, WT1, CDKN2A or CDKN2B, or RUNX1), and 1 of the 7 patients had monosomy 7. Of the 5 patients with MDS with excess blasts or MDS with unilineage dysplasia who underwent allogeneic hematopoietic stem-cell transplantation (HSCT), 4 patients remain free of MDS without recurrence of symptoms of cerebral adrenoleukodystrophy, and 1 patient died from presumed graft-versus-host disease 20 months after HSCT (49 months after receiving eli-cel). The patient with AML is alive and had full donor chimerism after HSCT; the patient with the most recent case of MDS is alive and awaiting HSCT. CONCLUSIONS: Hematologic cancer developed in a subgroup of patients who were treated with eli-cel; the cases are associated with clonal vector insertions within oncogenes and clonal evolution with acquisition of somatic genetic defects. (Funded by Bluebird Bio; ALD-102, ALD-104, and LTF-304 ClinicalTrials.gov numbers, NCT01896102, NCT03852498, and NCT02698579, respectively.).


Sujet(s)
Adrénoleucodystrophie , Thérapie génétique , Vecteurs génétiques , Tumeurs hématologiques , Lentivirus , Adolescent , Enfant , Femelle , Humains , Mâle , Adrénoleucodystrophie/thérapie , Adrénoleucodystrophie/génétique , Membre-1 de la sous-famille D de transporteurs à cassette liant l'ATP/génétique , Évolution clonale/génétique , Études de suivi , Thérapie génétique/effets indésirables , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/effets indésirables , Tumeurs hématologiques/épidémiologie , Tumeurs hématologiques/génétique , Lentivirus/génétique , Syndromes myélodysplasiques/épidémiologie , Syndromes myélodysplasiques/génétique
4.
N Engl J Med ; 391(14): 1302-1312, 2024 Oct 10.
Article de Anglais | MEDLINE | ID: mdl-39383459

RÉSUMÉ

BACKGROUND: Cerebral adrenoleukodystrophy is a severe form of X-linked adrenoleukodystrophy characterized by white-matter disease, loss of neurologic function, and early death. Elivaldogene autotemcel (eli-cel) gene therapy, which consists of autologous CD34+ cells transduced with Lenti-D lentiviral vector containing ABCD1 complementary DNA, is being tested in persons with cerebral adrenoleukodystrophy. METHODS: In a phase 2-3 study, we evaluated the efficacy and safety of eli-cel therapy in boys with early-stage cerebral adrenoleukodystrophy and evidence of active inflammation on magnetic resonance imaging (MRI). The primary efficacy end point was survival without any of six major functional disabilities at month 24. The secondary end points included overall survival at month 24 and the change from baseline to month 24 in the total neurologic function score. RESULTS: A total of 32 patients received eli-cel; 29 patients (91%) completed the 24-month study and are being monitored in the long-term follow-up study. At month 24, none of these 29 patients had major functional disabilities; overall survival was 94%. At the most recent assessment (median follow-up, 6 years), the neurologic function score was stable as compared with the baseline score in 30 of 32 patients (94%); 26 patients (81%) had no major functional disabilities. Four patients had adverse events that were directly related to eli-cel. Myelodysplastic syndrome (MDS) with excess blasts developed in 1 patient at month 92; the patient underwent allogeneic hematopoietic stem-cell transplantation and did not have MDS at the most recent follow-up. CONCLUSIONS: At a median follow-up of 6 years after lentiviral gene therapy, most patients with early cerebral adrenoleukodystrophy and MRI abnormalities had no major functional disabilities. However, insertional oncogenesis is an ongoing risk associated with the integration of viral vectors. (Funded by Bluebird Bio; ALD-102 and LTF-304 ClinicalTrials.gov numbers NCT01896102 and NCT02698579, respectively.).


Sujet(s)
Membre-1 de la sous-famille D de transporteurs à cassette liant l'ATP , Adrénoleucodystrophie , Thérapie génétique , Vecteurs génétiques , Lentivirus , Adolescent , Enfant , Enfant d'âge préscolaire , Humains , Mâle , Adrénoleucodystrophie/diagnostic , Adrénoleucodystrophie/génétique , Adrénoleucodystrophie/mortalité , Adrénoleucodystrophie/thérapie , Membre-1 de la sous-famille D de transporteurs à cassette liant l'ATP/génétique , Encéphale/imagerie diagnostique , Encéphale/anatomopathologie , Thérapie génétique/effets indésirables , Thérapie génétique/méthodes , Vecteurs génétiques/administration et posologie , Vecteurs génétiques/effets indésirables , Transplantation de cellules souches hématopoïétiques , Lentivirus/génétique , Imagerie par résonance magnétique , Études de suivi , Résultat thérapeutique , Syndromes myélodysplasiques/épidémiologie , Syndromes myélodysplasiques/génétique
6.
Clin Lab ; 70(10)2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39382936

RÉSUMÉ

BACKGROUND: The aim of the study was to improve the clinical cognition of nonaccelerating myelodysplastic/myeloproliferative neoplasms-unclassifiable (MDS/MPN-U) with 5q- karyotype and to avoid misdiagnosis or delayed diagnosis. METHODS: The clinical manifestations and laboratory results of a patient with nonaccelerating MDS/MPN-U with 5q- karyotype were analyzed, and related literature was reviewed. RESULTS: The patient was admitted to hospital mainly due to chest tightness and shortness of breath, aggravated for 4 days. After admission, combined with clinical manifestations, bone marrow cell morphology, immunology, multiparameter flow cytometry, cytogenetics and molecular biology, etc., the final diagnosis was MDS-MPN-U. CONCLUSIONS: Research on the correlation between MPN-U and MDS with 5q deletion is still needed. Clinically, MPN-U combined with MDS is prone to misdiagnosis. In diagnosing MPN-U patients, it is essential to not only complete routine and immunological tests but also consider clinical manifestations and laboratory results. It is crucial to be vigilant about the possibility of concurrent diseases, especially cancer, and to choose targeted examinations in a timely manner to avoid missed or incorrect diagnoses.


Sujet(s)
Délétion de segment de chromosome , Chromosomes humains de la paire 5 , Humains , Chromosomes humains de la paire 5/génétique , Mâle , Syndromes myélodysplasiques/diagnostic , Syndromes myélodysplasiques/génétique , Maladies myélodysplasiques-myéloprolifératives/diagnostic , Maladies myélodysplasiques-myéloprolifératives/génétique , Caryotype , Syndromes myéloprolifératifs/diagnostic , Syndromes myéloprolifératifs/génétique , Caryotypage , Sujet âgé
7.
J Cell Mol Med ; 28(18): e70078, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39334509

RÉSUMÉ

Myelodysplastic syndromes (MDS) are myeloid malignancies with heterogeneous genotypes and phenotypes, characterized by ineffective haematopoiesis and a high risk of progression towards acute myeloid leukaemia (AML). Prognosis for patients treated with hypomethylating agents (HMAs), as is azacytidine, the main drug used as frontline therapy for MDS is mostly based on cytogenetics and next generation sequencing (NGS) of the initial myeloid clone. Although the critical influence of the epigenetic landscape upon cancer cells survival and development as well on tumour environment establishment is currently recognized and approached within current clinical practice in MDS, the heterogenous response of the patients to epigenetic therapy is suggesting a more complex mechanism of action, as is the case of RNA methylation. In this sense, the newly emerging field of epitranscriptomics could provide a more comprehensive perspective upon the modulation of gene expression in malignancies, as is the proof-of-concept of MDS. We initially did RNA methylation sequencing on MDS patients (n = 6) treated with azacytidine and compared responders with non-responders. Afterwards, the genes identified were assessed in vitro and afterwards validated on a larger cohort of MDS patients treated with azacytidine (n = 58). Our data show that a more accurate prognosis could be based on analysing the methylome and thus we used methylation sequencing to differentially split high-grade MDS patients with identical demographical and cytogenetic features, between azacytidine responders and non-responders.


Sujet(s)
Azacitidine , Méthylation de l'ADN , Syndromes myélodysplasiques , Humains , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/anatomopathologie , Azacitidine/pharmacologie , Azacitidine/usage thérapeutique , Femelle , Sujet âgé , Mâle , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Adulte d'âge moyen , Transcriptome/génétique , Transcriptome/effets des médicaments et des substances chimiques , Sujet âgé de 80 ans ou plus , Épigenèse génétique/effets des médicaments et des substances chimiques , Analyse de séquence d'ARN , Antimétabolites antinéoplasiques/usage thérapeutique , Antimétabolites antinéoplasiques/pharmacologie , Pronostic , Séquençage nucléotidique à haut débit , Analyse de profil d'expression de gènes , RNA Methylation
8.
Int J Mol Sci ; 25(18)2024 Sep 23.
Article de Anglais | MEDLINE | ID: mdl-39337700

RÉSUMÉ

The accurate diagnosis and classification of myelodysplastic/myeloproliferative neoplasm (MDS/MPN) are challenging due to the overlapping pathological and molecular features of myelodysplastic syndrome (MDS) and myeloproliferative neoplasm (MPN). We investigated the genomic landscape in different MDS/MPN subtypes, including chronic myelomonocytic leukemia (CMML; n = 97), atypical chronic myeloid leukemia (aCML; n = 8), MDS/MPN-unclassified (MDS/MPN-U; n = 44), and MDS/MPN with ring sideroblasts and thrombocytosis (MDS/MPN-RS-T; n = 12). Our study indicated that MDS/MPN is characterized by mutations commonly identified in myeloid neoplasms, with TET2 (52%) being the most frequently mutated gene, followed by ASXL1 (38.7%), SRSF2 (34.7%), and JAK2 (19.7%), among others. However, the distribution of recurrent mutations differs across the MDS/MPN subtypes. We confirmed that specific gene combinations correlate with specific MDS/MPN subtypes (e.g., TET2/SRSF2 in CMML, ASXL1/SETBP1 in aCML, and SF3B1/JAK2 in MDS/MPN-RS-T), with MDS/MPN-U being the most heterogeneous. Furthermore, we found that older age (≥65 years) and mutations in RUNX1 and TP53 were associated with poorer clinical outcomes in CMML (p < 0.05) by multivariate analysis. In MDS/MPN-U, CBL mutations (p < 0.05) were the sole negative prognostic factors identified in our study by multivariate analysis (p < 0.05). Overall, our study provides genetic insights into various MDS/MPN subtypes, which may aid in diagnosis and clinical decision-making for patients with MDS/MPN.


Sujet(s)
Mutation , Maladies myélodysplasiques-myéloprolifératives , Protéines proto-oncogènes , Humains , Sujet âgé , Mâle , Femelle , Adulte d'âge moyen , Maladies myélodysplasiques-myéloprolifératives/génétique , Maladies myélodysplasiques-myéloprolifératives/classification , Sujet âgé de 80 ans ou plus , Protéines proto-oncogènes/génétique , Adulte , Dioxygenases , Facteurs d'épissage riches en sérine-arginine/génétique , Kinase Janus-2/génétique , Protéines de liaison à l'ADN/génétique , Génomique/méthodes , Leucémie myélomonocytaire chronique/génétique , Leucémie myélomonocytaire chronique/anatomopathologie , Syndromes myélodysplasiques/génétique , Sous-unité alpha 2 du facteur CBF/génétique , Syndromes myéloprolifératifs/génétique , Syndromes myéloprolifératifs/diagnostic , Protéines de répression/génétique , Facteurs d'épissage des ARN/génétique , Protéines de transport , Protéines nucléaires
9.
Mol Biol Rep ; 51(1): 985, 2024 Sep 15.
Article de Anglais | MEDLINE | ID: mdl-39278886

RÉSUMÉ

OBJECTIVE: To evaluate the frequency and prognostic significance of DTA (DNMT3A、TET2、ASXL1) gene mutation and co-occurring mutations in patients with myelodysplastic syndrome (MDS). METHODS: The clinical data of 102 newly diagnosed MDS patients who accepted Next Generation Sequencing (NGS) was retrospectively analyzed. According to whether the patients had DTA gene mutation, the patients were divided into DTA mutated (DTA-mut) group and wild type (DTA-wt) group, and the relationship between gene mutation and clinical characteristics and prognosis was analyzed. RESULTS: Among the 102 MDS patients, 96% (98/102) presented with mutation, while the mean number of mutations was 3.04 mutations/patient. DTA-mut was detected in 56.9% (58/102) patients. The most frequent co-mutated genes in DTA-mut group were SF3B1 (25.8%), RUNX1 (24.1%), U2AF1 (18.9%), SRSF2, EZH2, SETBP1 (17.2%), STAG2 (15.5%), IDH2 (12.1%) and BCOR, CBL (10.3%). The two groups showed no significant differences in ages, blood parameters, bone marrow blasts, WHO 2022 classification, IPSS-R risk category and rate of conversion to leukemia. Compared with the DTA-wt group, the mutation frequency of RUNX1 was higher (P = 0.02), while mutation frequency of TP53 was lower (P = 0.001) and the mutation frequency of ≥ 3 co-mutated genes was higher in DTA-mut group (P = 0.00). Survival analysis showed that the overall survivals (OS) of DTA-mut patients was significantly inferior to that of DTA-wt patients (P = 0.0332). According to IPSS-R classification, a statistically significant difference in OS was only observed in higher risk (IPSS-R > 3.5) group (P = 0.0058). In the context of DTA mutation, the OS of patients with RUNX1 mutation was shorter than that of patients without RUNX1 mutation significantly (P = 0.0074). The OS of patients with SF3B1 mutation was longer than that of patients without SF3B1 mutation, but there was no statistical difference (P = 0.0827). DTA mutations were not independent prognostic factors when DTA and co-mutated genes with frequency > 10% were considered in Cox regression model (P = 0.329). However, multivariate analysis confirmed an independently adverse prognosis of RUNX1 co-mutation (P = 0.042, HR = 2.426, 95% CI:1.031-5.711) in DTA-mut cohort. Moreover, our multivariable analysis suggests that SRSF2-mut was an independent poor prognostic factor for all MDS patients (P = 0.047), but lost significance (P = 0.103) for DTA-mut patients. CONCLUSIONS: DTA mutations are frequently observed in patients with MDS, often accompanied by genes involved in RNA splicing and transcription factors like SF3B1 and RUNX1. DTA and concomitant mutations affect prognosis in MDS patients and RUNX1 was identified as an independent poor prognostic factor in patients with DTA mutations.


Sujet(s)
DNA (cytosine-5-)-methyltransferase , DNA methyltransferase 3A , Protéines de liaison à l'ADN , Dioxygenases , Mutation , Syndromes myélodysplasiques , Protéines proto-oncogènes , Protéines de répression , Humains , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/mortalité , Mâle , Femelle , Mutation/génétique , Adulte d'âge moyen , Pronostic , Sujet âgé , Adulte , Protéines proto-oncogènes/génétique , DNA (cytosine-5-)-methyltransferase/génétique , Protéines de répression/génétique , Protéines de liaison à l'ADN/génétique , Études rétrospectives , Sujet âgé de 80 ans ou plus , Séquençage nucléotidique à haut débit/méthodes , Adolescent , Jeune adulte
10.
Elife ; 132024 Sep 05.
Article de Anglais | MEDLINE | ID: mdl-39235452

RÉSUMÉ

Mutational profiles of myelodysplastic syndromes (MDS) have established that a relatively small number of genetic aberrations, including SF3B1 and SRSF2 spliceosome mutations, lead to specific phenotypes and prognostic subgrouping. We performed a multi-omics factor analysis (MOFA) on two published MDS cohorts of bone marrow mononuclear cells (BMMNCs) and CD34 + cells with three data modalities (clinical, genotype, and transcriptomics). Seven different views, including immune profile, inflammation/aging, retrotransposon (RTE) expression, and cell-type composition, were derived from these modalities to identify the latent factors with significant impact on MDS prognosis. SF3B1 was the only mutation among 13 mutations in the BMMNC cohort, indicating a significant association with high inflammation. This trend was also observed to a lesser extent in the CD34 + cohort. Interestingly, the MOFA factor representing the inflammation shows a good prognosis for MDS patients with high inflammation. In contrast, SRSF2 mutant cases show a granulocyte-monocyte progenitor (GMP) pattern and high levels of senescence, immunosenescence, and malignant myeloid cells, consistent with their poor prognosis. Furthermore, MOFA identified RTE expression as a risk factor for MDS. This work elucidates the efficacy of our integrative approach to assess the MDS risk that goes beyond all the scoring systems described thus far for MDS.


Sujet(s)
Inflammation , Syndromes myélodysplasiques , Syndromes myélodysplasiques/immunologie , Syndromes myélodysplasiques/génétique , Humains , Pronostic , Inflammation/génétique , Inflammation/immunologie , Facteurs d'épissage riches en sérine-arginine/génétique , Facteurs d'épissage riches en sérine-arginine/métabolisme , Mutation , Facteurs d'épissage des ARN/génétique , Facteurs d'épissage des ARN/métabolisme , Moelle osseuse/immunologie , Études de cohortes , Rétroéléments/génétique
11.
Am J Dermatopathol ; 46(10): 637-647, 2024 Oct 01.
Article de Anglais | MEDLINE | ID: mdl-39293663

RÉSUMÉ

ABSTRACT: We present the histopathology of 12 skin biopsies from 6 patients with vacuoles, enzyme E1, X-linked, autoinflammatory, somatic syndrome and review the literature. The age of these 6 men ranges from 62 to 83 years (median of 70 years). UBA1 mutation was documented in all 6 patients. Multiple organ systems were involved with constitutional symptoms noted in 4 of 6 patients (67%), cutaneous involvement in 6 of 6 patients (100%), hematologic abnormalities in 6 of 6 patients (100%), pulmonary involvement in 4 of 6 patients (67%), musculoskeletal abnormalities in 3 of 6 patients (50%), vascular thrombosis in 2 of 6 patients (33%), ocular involvement in 2 of 6 patients (33%), and gastrointestinal involvement in 5 of 6 patients (83%). Of the 6 presented patients, neutrophilic dermatosis was seen in 3 biopsies, histiocytoid neutrophilic dermatosis in 1 biopsy, neutrophilic dermatosis with vasculitis in 1 biopsy, neutrophilic and granulomatous dermatitis in 2 biopsies, septal panniculitis consistent with erythema nodosum in 2 biopsies, and nonspecific patterns in 3 biopsies. In summary, neutrophilic dermatosis, small-vessel vasculitis, and panniculitis are frequent histopathologic patterns noted in decreasing frequency in skin biopsies of the patients with vacuoles, enzyme E1, X-linked, autoinflammatory, somatic syndrome. However, the histopathologic findings can be diverse, nonspecific in some instances, and varied among different biopsies obtained from the same patient.


Sujet(s)
Syndromes myélodysplasiques , Maladies génétiques de la peau , Peau , Sujet âgé , Sujet âgé de 80 ans ou plus , Humains , Mâle , Adulte d'âge moyen , Biopsie , Mutation , Peau/anatomopathologie , Syndrome , Ubiquitin-activating enzymes/génétique , Maladies génétiques de la peau/diagnostic , Maladies génétiques de la peau/génétique , Maladies génétiques de la peau/anatomopathologie , Syndromes myélodysplasiques/diagnostic , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/anatomopathologie
12.
Zhonghua Xue Ye Xue Za Zhi ; 45(7): 651-659, 2024 Jul 14.
Article de Chinois | MEDLINE | ID: mdl-39231769

RÉSUMÉ

Objective: To analyze the clinical characteristics and prognosis of patients with myelodysplastic syndrome (MDS) with a bone marrow nucleated erythroid cell proportion of greater than or equal to 50% (MDS-E) . Methods: The clinical characteristics and prognostic factors of patients with MDS-E were retrospectively analyzed by collecting the case data of 1 436 newly treated patients with MDS diagnosed in the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences from May 2014 to June 2023. Results: A total of 1 436 newly diagnosed patients with complete data were included in the study, of which 337 (23.5%) patients with MDS-E had a younger age of onset and lower neutrophil and platelet counts compared with those in patients with an erythroid cell proportion of less than 50% (MDS-NE) (all P<0.05). The proportion of MDS cases with ring sideroblasts (MDS-RS) was higher in the MDS-E group than in the MDS-NE group, and multi-hit TP53 mutations were more enriched in the MDS-E group than in the MDS-NE group (all P<0.05). Among patients with MDS-RS, the frequency of complex karyotypes and the TP53 mutation rate were significantly lower in the MDS-E group than in the MDS-NE group (0 vs 11.9%, P=0.048 and 2.4% vs 15.1%, P=0.053, respectively). Among patients with TP53 mutations, the frequencies of complex karyotypes and multi-hit TP53 mutations were significantly higher in the MDS-E group than in the MDS-NE group (87.5% vs 64.6%, P=0.003 and 84.0% vs 54.2%, P<0.001, respectively). Survival analysis of patients with MDS-RS found that the overall survival (OS) in the MDS-E group was better than that in the MDS-NE group [not reached vs 63 (95% CI 53.3-72.7) months, P=0.029]. Among patients with TP53 mutations and excess blasts, the OS in the MDS-E group was worse than that in the MDS-NE group [6 (95% CI 2.2-9.8) months vs 12 (95% CI 8.9-15.1) months, P=0.022]. Multivariate analysis showed that age of ≥65 years (HR=2.47, 95% CI 1.43-4.26, P=0.001), mean corpuscular volume (MCV) of ≤100 fl (HR=2.62, 95% CI 1.54-4.47, P<0.001), and TP53 mutation (HR=2.31, 95% CI 1.29-4.12, P=0.005) were poor prognostic factors independent of the Revised International Prognostic Scoring System (IPSS-R) prognosis stratification in patients with MDS-E. Conclusion: Among patients with MDS-RS, MDS-E was strongly associated with a lower proportion of complex karyotypes and TP53 mutations, and the OS in the MDS-E group was longer than that in the MDS-NE group. Among patients with TP53 mutations, MDS-E was strongly associated with complex karyotypes and multi-hit TP53 mutations, and among TP53-mutated patients with excess blasts, the OS in the MDS-E group was shorter than that in the MDS-NE group. Age of ≥65 years, MCV of ≤100 fl, and TP53 mutation were independent adverse prognostic factors affecting OS in patients with MDS-E.


Sujet(s)
Mutation , Syndromes myélodysplasiques , Humains , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/diagnostic , Pronostic , Études rétrospectives , Moelle osseuse/anatomopathologie , Cellules de la moelle osseuse , Mâle , Femelle , Taux de survie , Protéine p53 suppresseur de tumeur/génétique , Adulte d'âge moyen
13.
Nat Commun ; 15(1): 7360, 2024 Aug 28.
Article de Anglais | MEDLINE | ID: mdl-39198401

RÉSUMÉ

Hypomethylating agents (HMAs) are frontline therapies for Myelodysplastic Neoplasms (MDS) and Acute Myeloid Leukemia (AML). However, acquired resistance and treatment failure are commonplace. To address this, we perform a genome-wide CRISPR-Cas9 screen in a human MDS-derived cell line, MDS-L, and identify TOPORS as a loss-of-function target that synergizes with HMAs, reducing leukemic burden and improving survival in xenograft models. We demonstrate that depletion of TOPORS mediates sensitivity to HMAs by predisposing leukemic blasts to an impaired DNA damage response (DDR) accompanied by an accumulation of SUMOylated DNMT1 in HMA-treated TOPORS-depleted cells. The combination of HMAs with targeting of TOPORS does not impair healthy hematopoiesis. While inhibitors of TOPORS are unavailable, we show that inhibition of protein SUMOylation with TAK-981 partially phenocopies HMA-sensitivity and DDR impairment. Overall, our data suggest that the combination of HMAs with inhibition of SUMOylation or TOPORS is a rational treatment option for High-Risk MDS (HR-MDS) or AML.


Sujet(s)
Systèmes CRISPR-Cas , Résistance aux médicaments antinéoplasiques , Leucémie aigüe myéloïde , Syndromes myélodysplasiques , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/métabolisme , Animaux , Résistance aux médicaments antinéoplasiques/génétique , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Lignée cellulaire tumorale , Souris , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/anatomopathologie , Syndromes myélodysplasiques/métabolisme , Sumoylation/effets des médicaments et des substances chimiques , Ubiquitin-protein ligases/métabolisme , Ubiquitin-protein ligases/génétique , Altération de l'ADN/effets des médicaments et des substances chimiques , Méthylation de l'ADN/effets des médicaments et des substances chimiques , Tests d'activité antitumorale sur modèle de xénogreffe , DNA (Cytosine-5-)-methyltransferase 1/métabolisme , DNA (Cytosine-5-)-methyltransferase 1/génétique , DNA (Cytosine-5-)-methyltransferase 1/antagonistes et inhibiteurs , Femelle
14.
PLoS One ; 19(8): e0309456, 2024.
Article de Anglais | MEDLINE | ID: mdl-39186541

RÉSUMÉ

The metabolomic landscape in myelodysplastic syndrome (MDS) is highly deregulated and presents promising avenues for understanding disease pathogenesis and potential molecular dependencies. Here, we evaluated the transcriptomic landscape in MDS in multiple independent studies focusing more on metabolomics pathways. Identifying molecular dependencies will pave the way for a more precise disease stratification as well as the development of novel personalized treatment strategies. The study adopted a retrospective, cross-sectional approach, utilizing transcriptomic data from multiple MDS studies. The transcriptomic data were then subjected to comprehensive analyses, including differential gene expression, gene enrichment analysis, gene co-expression analysis, protein-protein interaction analyses, and survival analyses. PSAT1 showed a significant upregulation profile in MDS patients. This observed upregulation is correlated with the deregulation of immune-related pathways in MDS samples. This observation suggests a novel role for PSAT1 in immune modulation and potentially in augmenting immune evasion, which may lead to poor prognosis. This was evident in other tumors in the TCGA database, where cancer patients with high PSAT1 expression have a shorter overall survival. This study unveils a novel potential therapeutic avenue in MDS. Identifying the role of the PSAT1 gene sheds light on the disease's intricate biology, highlighting the ongoing cross-talk between metabolism and immune regulation, which may pave the way for innovative treatment modalities.


Sujet(s)
Syndromes myélodysplasiques , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Syndromes myélodysplasiques/thérapie , Humains , Études rétrospectives , Études transversales , Pronostic , Transcriptome , Cartes d'interactions protéiques , Analyse de profil d'expression de gènes , Mâle , Femelle
15.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 32(4): 1173-1180, 2024 Aug.
Article de Chinois | MEDLINE | ID: mdl-39192415

RÉSUMÉ

OBJECTIVE: To investigate the clinical characteristics and survival analysis of myelodysplastic syndromes (MDS) with RUNX1 gene mutation. METHODS: Clinical data of 177 newly diagnosed MDS patients admitted to the Department of Hematology, the Second Affiliated Hospital of Air Force Military Medical University from October 1, 2015 to October 31, 2022 were retrospectively analyzed. Gene mutation detection was performed by second-generation sequencing technology, and clinical characteristics and prognosis of patients with RUNX1 gene mutation were analyzed. RESULTS: A total of 30 cases (16.95%) of RUNX1 gene mutations were detected, including 15 missense mutations (50.0%), 9 frameshift deletion mutations (30.0%), 4 splice site mutations (13.3%), 1 insertion mutation (3.3%), and 1 nonsense mutation (3.3%). Patients with RUNX1 mutations had a median age of 68.5 years at diagnosis (range: 62.25-78.50 years old). There were no significantly differences between RUNX1 mutations and wild type patients in age distribution, gender, peripheral blood white blood cell count, hemoglobin level, bone marrow and peripheral blood blasts ratio, IPSS-R cytogenetics, IPSS-R stage, etc. (P >0.05). However, there were statistically significant differences in platelet count and whether complicated karyotype. Compared with patients without RUNX1 gene mutation, patients with RUNX1 gene mutation had lower platelet count (P =0.018), and were less likely to have complicatedkaryotype at initial diagnosis (P =0.01). Cox proportional hazards model analysis showed that when other covariates remained unchanged, the higher the platelet count, the better the survival of patients (HR=0.995, 95%CI : 0.990-0.999, P =0.036); In the IPSS-M prognostic stratification, keeping other covariates unchanged, the risk of progression or death of myelodysplastic syndrome was significantly lower in the medium to high-risk and low-risk groups compared with the high-risk group (HR=0.149, 95%CI : 0.031-0.721, P =0.018; HR=0.026, 95%CI : 0.003-0.234, P =0.001). Survival analysis showed that MDS patients with RUNX1 gene mutation had worse overall survival time (P < 0.001). Patients with RUNX1 mutation had worse OS than non-mutation patients in the early WHO group. RUNX1 mutation and IPSS-M risk stratification mean OS and mean LFS were worse in low-risk patients than in non-mutated patients. CONCLUSION: RUNX1 gene mutation is an adverse prognostic factor in MDS patients, especially in the IPSS-M prognosis stratification group of low-risk, medium-low risk, medium-high risk and WHO classification of early patients.


Sujet(s)
Sous-unité alpha 2 du facteur CBF , Mutation , Syndromes myélodysplasiques , Humains , Sous-unité alpha 2 du facteur CBF/génétique , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/diagnostic , Pronostic , Adulte d'âge moyen , Sujet âgé , Études rétrospectives , Mâle , Femelle
16.
Nat Commun ; 15(1): 7138, 2024 Aug 20.
Article de Anglais | MEDLINE | ID: mdl-39164231

RÉSUMÉ

Telomere shortening is a prominent hallmark of aging and is emerging as a characteristic feature of Myelodysplastic Syndromes (MDS) and Idiopathic Pulmonary Fibrosis (IPF). Optimal telomerase activity prevents progressive shortening of telomeres that triggers DNA damage responses. However, the upstream regulation of telomerase holoenzyme components remains poorly defined. Here, we identify RIOK2, a master regulator of human blood cell development, as a critical transcription factor for telomere maintenance. Mechanistically, loss of RIOK2 or its DNA-binding/transactivation properties downregulates mRNA expression of both TRiC and dyskerin complex subunits that impairs telomerase activity, thereby causing telomere shortening. We further show that RIOK2 expression is diminished in aged individuals and IPF patients, and it strongly correlates with shortened telomeres in MDS patient-derived bone marrow cells. Importantly, ectopic expression of RIOK2 alleviates telomere shortening in IPF patient-derived primary lung fibroblasts. Hence, increasing RIOK2 levels prevents telomere shortening, thus offering therapeutic strategies for telomere biology disorders.


Sujet(s)
Protéines du cycle cellulaire , Fibrose pulmonaire idiopathique , Protéines nucléaires , Telomerase , Raccourcissement des télomères , Humains , Fibrose pulmonaire idiopathique/génétique , Fibrose pulmonaire idiopathique/métabolisme , Fibrose pulmonaire idiopathique/anatomopathologie , Telomerase/métabolisme , Telomerase/génétique , Protéines nucléaires/métabolisme , Protéines nucléaires/génétique , Protéines du cycle cellulaire/métabolisme , Protéines du cycle cellulaire/génétique , Fibroblastes/métabolisme , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Télomère/métabolisme , Télomère/génétique , Régulation de l'expression des gènes , Poumon/métabolisme , Poumon/anatomopathologie
17.
Cancer Biomark ; 41(1): 55-68, 2024.
Article de Anglais | MEDLINE | ID: mdl-39213050

RÉSUMÉ

BACKGROUND: Myelodysplastic syndrome (MDS) features bone marrow failure and a heightened risk of evolving into acute myeloid leukemia (AML), increasing with age and reducing overall survival. Given the unfavorable outcomes of MDS, alternative treatments are necessary. Glutamine, the most abundant amino acid in the blood, is metabolized first by the enzyme glutaminase (GLS). OBJECTIVES: To investigate whether GLS is involved in the progression of MDS. The efficacy of GLS inhibitors (CB839 or IPN60090) and BCL2 inhibitor venetoclax was also examined. METHODS: We employed GLS inhibitors (CB839, IPN60090) and the BCL2 inhibitor venetoclax, prepared as detailed. MDS and AML cell lines were cultured under standard and modified (hypoxic, glutamine-free) conditions. Viability, proliferation, and caspase activity were assessed with commercial kits. RT-PCR quantified gene expression post-shRNA transfection. Mitochondrial potential, ATP levels, proteasome activity, and metabolic functions were evaluated using specific assays. Statistical analyses (t-tests, ANOVA) validated the findings. RESULTS: The glutamine-free medium inhibited the growth of MDS cells. GLS1 expression was higher in AML cells than in normal control samples (GSE15061), whereas GLS2 expression was not. Treatment of MDS and AML cells for 72 h was inhibited in a dose-dependent manner by GLS inhibitors. Co-treatment with the B-cell lymphoma 2 (BCL2) inhibitor venetoclax and GLS inhibitors increased potency. Cells transfected with GLS1 short hairpin RNA showed suppressed proliferation under hypoxic conditions and increased sensitivity to venetoclax. CONCLUSIONS: Targeting glutaminolysis and BCL2 inhibition enhances the therapeutic efficacy and has been proposed as a novel strategy for treating high-risk MDS and AML.


Sujet(s)
Composés hétérocycliques bicycliques , Glutaminase , Leucémie aigüe myéloïde , Syndromes myélodysplasiques , Sulfonamides , Thiadiazoles , Glutaminase/antagonistes et inhibiteurs , Glutaminase/métabolisme , Glutaminase/génétique , Humains , Leucémie aigüe myéloïde/traitement médicamenteux , Leucémie aigüe myéloïde/anatomopathologie , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/métabolisme , Syndromes myélodysplasiques/traitement médicamenteux , Syndromes myélodysplasiques/anatomopathologie , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/métabolisme , Composés hétérocycliques bicycliques/pharmacologie , Composés hétérocycliques bicycliques/usage thérapeutique , Sulfonamides/pharmacologie , Thiadiazoles/pharmacologie , Lignée cellulaire tumorale , Prolifération cellulaire/effets des médicaments et des substances chimiques , Protéines proto-oncogènes c-bcl-2/métabolisme , Protéines proto-oncogènes c-bcl-2/génétique , Benzèneacétamides/pharmacologie , Composés benzylidéniques/pharmacologie , Apoptose/effets des médicaments et des substances chimiques , Sulfures
18.
Int J Hematol ; 120(4): 417-426, 2024 Oct.
Article de Anglais | MEDLINE | ID: mdl-39150677

RÉSUMÉ

Germline (GL) predisposition to acute myeloid leukemia (AML) has been established as an independent disease entity in the latest World Health Organization classification. Following the American College of Medical Genetics and Genomics guidelines, GL variants were interpreted as causal if they were classified as "pathogenic." GL predisposition can be divided into three groups with different phenotypes, and play an important role in the pathogenesis of adult-onset AML. The clinical course and age of onset of myeloid neoplasms varied considerably for each gene. For example, patients with GATA2 GL variants develop AML before the age of 30 along with bone marrow failure, whereas those with DDX41 GL variants tend to develop AML after the age of 50 without any preceding hematological abnormalities or organ dysfunction. A comprehensive analysis of adult-onset myelodysplastic syndromes in transplant donors showed a 7% frequency of pathogenic GL variants, with DDX41 being the most frequent gene mutation at approximately 3.8%. Future research on GL predisposition at any age of myeloid neoplasm onset will assist in early and accurate diagnosis, development of effective treatment strategies, and selection of suitable donors for stem cell transplantation.


Sujet(s)
DEAD-box RNA helicases , Prédisposition génétique à une maladie , Mutation germinale , Leucémie aigüe myéloïde , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/thérapie , Leucémie aigüe myéloïde/diagnostic , DEAD-box RNA helicases/génétique , Facteur de transcription GATA-2/génétique , Adulte , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/diagnostic , Âge de début
19.
Int J Mol Sci ; 25(16)2024 Aug 08.
Article de Anglais | MEDLINE | ID: mdl-39201354

RÉSUMÉ

Two new diagnostic classifications of acute myeloid leukemia (AML) were published in 2022 to update current knowledge on disease biology. In previous 2017-edition categories of AML with myelodysplasia-related changes, AML was not otherwise specified, but AML with mutated RUNX1 experienced profound changes. We performed whole exome sequencing on a cohort of 69 patients with cytogenetic intermediate-risk AML that belonged to these diagnostic categories to correlate their mutational pattern and copy-number alterations with their new diagnostic distribution. Our results show that 45% of patients changed their diagnostic category, being AML myelodysplasia-related the most enlarged, mainly due to a high frequency of myelodysplasia-related mutations (58% of patients). These showed a good correlation with multilineage dysplasia and/or myelodysplastic syndrome history, but at the same time, 21% of de novo patients without dysplasia also presented them. RUNX1 was the most frequently mutated gene, with a high co-occurrence rate with other myelodysplasia-related mutations. We found a high prevalence of copy-neutral loss of heterozygosity, frequently inducing a homozygous state in particular mutated genes. Mild differences in current classifications explain the diagnostic disparity in 10% of patients, claiming a forthcoming unified classification.


Sujet(s)
Sous-unité alpha 2 du facteur CBF , Exome Sequencing , Leucémie aigüe myéloïde , Mutation , Humains , Leucémie aigüe myéloïde/génétique , Leucémie aigüe myéloïde/diagnostic , Femelle , Mâle , Adulte d'âge moyen , Sujet âgé , Adulte , Sous-unité alpha 2 du facteur CBF/génétique , Variations de nombre de copies de segment d'ADN , Sujet âgé de 80 ans ou plus , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/diagnostic , Syndromes myélodysplasiques/classification
20.
Leuk Res ; 144: 107563, 2024 Sep.
Article de Anglais | MEDLINE | ID: mdl-39178611

RÉSUMÉ

Myelodysplastic syndrome, or myelodysplastic neoplasms, are a rare finding in pediatric, adolescent, and young adult (AYA) patients. More literature is needed to highlight trends of survival or treatment resistance in subpopulations to improve treatment. Here we report a single center retrospective analysis of pediatric and AYA patients from 2000 to 2022 including molecular and cytogenetic data. Using the IPSS-R and IPSS-M, which have been reported exclusively in adults, and excluding patients with bone marrow failure syndromes, we analyzed 119 pediatric and AYA patients with myelodysplastic neoplasms. Therapy-related myelodysplastic neoplasms were present in 36 % of patients, and 31 % of patients developed acute myeloid leukemia. The 5-year overall survival (OS) rate for the entire cohort was 45 %. Contrary to young adults and older adults, mutations were not common in pediatrics. Those who underwent stem cell transplant (SCT)(at any time) had significantly longer median OS. Although SCT at any time improved OS in the de novo myelodysplastic neoplasm group, the choice of the initial treatment with intensive chemotherapy, hypomethylating agents, or SCT did not significantly alter OS. Median OS was shorter in the pediatric group (<18 years old) and longer for those with isolated deletion of 5q or TET2 mutation, but these were not significant findings. Median OS was significantly shorter in those with monosomy 7 or 7q deletion and those with therapy-related myelodysplastic neoplasms. These findings build on previously reported findings and encourage the use of SCT along with molecular and cytogenetic analysis.


Sujet(s)
Syndromes myélodysplasiques , Humains , Adolescent , Études rétrospectives , Syndromes myélodysplasiques/thérapie , Syndromes myélodysplasiques/génétique , Syndromes myélodysplasiques/mortalité , Syndromes myélodysplasiques/étiologie , Mâle , Femelle , Jeune adulte , Enfant , Adulte , Enfant d'âge préscolaire , Mutation , Taux de survie , Résultat thérapeutique , Pronostic
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE