Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 38
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
2.
Am J Hematol ; 99(1): 68-78, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-37846894

RESUMEN

Contemporary risk models in primary myelofibrosis (PMF) include the mutation (MIPSS70) and mutation/karyotype enhanced (MIPSS70 plus/v2.0) international prognostic scoring systems. High molecular risk (HMR) mutations incorporated in one or both of these models include ASXL1, SRSF2, EZH2, IDH1/2, and U2AF1Q157; the current study examines additional prognostic contribution from more recently described HMR mutations, including CBL, NRAS, KRAS, RUNX1, and TP53. In a cohort of 363 informative cases (median age 58 years; 60% males), mutations included JAK2 61%, CALR 24%, MPL 6%, ASXL1 29%, SRSF2 10%, U2AF1Q157 5%, EZH2 10%, IDH1/2 4%, TP53 5%, CBL 5%, NRAS 7%, KRAS 4%, and RUNX1 4%. At a median follow-up of 4.6 years, 135 (37%) deaths and 42 (11.6%) leukemic transformations were recorded. Univariate analysis confirmed significant survival impact from the original MIPSS70/plus/v2.0 HMR mutations as well as CBL (HR 2.8; p < .001), NRAS (HR 2.4; p < .001), KRAS (HR 2.1; p = .01), and TP53 (HR 2.4; p = .004), but not RUNX1 mutations (HR 1.8; p = .08). Multivariate analysis (MVA) that included both the original and more recently described HMR mutations confirmed independent prognostic contribution from ASXL1 (HR 1.8; p = .007), SRSF2 (HR 4.3; p < .001), U2AF1Q157 (HR 2.9, p = .004), and EZH2 (HR 2.4; p < .001), but not from IDH1/2 (p = .3), TP53 (p = .2), CBL (p = .3), NRAS (p = .8) or KRAS (p = .2) mutations. The lack of additional prognostic value from CBL, NRAS, KRAS, RUNX1, and TP53 was further demonstrated in the setting of (i) MVA of mutations and karyotype, (ii) MVA of MIPSS70/plus/v2.0 composite scores and each one of the recently described HMR mutations, except TP53, and iii) modified MIPSS70/plus/plus v2.0 that included CBL, NRAS, KRAS, and TP53 as part of the HMR constituency, operationally referred to as "HMR+" category. Furthermore, "HMR+" enhancement of MIPSS70/plus/plus v2.0 did not result in improved model performance, as measured by C-statistics. We conclude that prognostic integrity of MIPSS70/plus/plus v2.0, as well as their genetic components, was sustained and their value not significantly upgraded by the inclusion of more recently described HMR mutations, including CBL, NRAS, KRAS, and RUNX1. Additional studies are needed to clarify the apparent additional prognostic value of TP53 mutation and its allelic state.


Asunto(s)
Subunidad alfa 2 del Factor de Unión al Sitio Principal , Mielofibrosis Primaria , Masculino , Humanos , Persona de Mediana Edad , Femenino , Pronóstico , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , Proteínas Proto-Oncogénicas p21(ras)/genética , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/genética , Mutación , Proteína p53 Supresora de Tumor/genética , Proteínas de la Membrana/genética , GTP Fosfohidrolasas/genética
3.
Int J Mol Sci ; 24(20)2023 Oct 18.
Artículo en Inglés | MEDLINE | ID: mdl-37895004

RESUMEN

The molecular basis of Down syndrome (DS) predisposition to leukemia is not fully understood but involves various factors such as chromosomal abnormalities, oncogenic mutations, epigenetic alterations, and changes in selection dynamics. Myeloid leukemia associated with DS (ML-DS) is preceded by a preleukemic phase called transient abnormal myelopoiesis driven by GATA1 gene mutations and progresses to ML-DS via additional mutations in cohesin genes, CTCF, RAS, or JAK/STAT pathway genes. DS-related ALL (ALL-DS) differs from non-DS ALL in terms of cytogenetic subgroups and genetic driver events, and the aberrant expression of CRLF2, JAK2 mutations, and RAS pathway-activating mutations are frequent in ALL-DS. Recent advancements in single-cell multi-omics technologies have provided unprecedented insights into the cellular and molecular heterogeneity of DS-associated hematologic neoplasms. Single-cell RNA sequencing and digital spatial profiling enable the identification of rare cell subpopulations, characterization of clonal evolution dynamics, and exploration of the tumor microenvironment's role. These approaches may help identify new druggable targets and tailor therapeutic interventions based on distinct molecular profiles, ultimately improving patient outcomes with the potential to guide personalized medicine approaches and the development of targeted therapies.


Asunto(s)
Síndrome de Down , Neoplasias Hematológicas , Humanos , Síndrome de Down/complicaciones , Síndrome de Down/genética , Síndrome de Down/patología , Quinasas Janus/metabolismo , Transducción de Señal/genética , Factores de Transcripción STAT/metabolismo , Mutación , Neoplasias Hematológicas/genética , Microambiente Tumoral
4.
Am J Hematol ; 97(7): 846-855, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-35338671

RESUMEN

Prognostic modeling in myelofibrosis (MF) has classically pursued the integration of informative clinical and hematological parameters to separate patients' categories with different outcomes. Modern stratification includes also genetic data from karyotype and mutations. However, some poorly standardized variables, as peripheral blood (PB) blast count by morphology, are still included. In this study, we used multiparameter flow cytometry (MFC) with the aim of improving performance of existing scores. We studied 363 MF patients with available MFC files for PB CD34+ cells count determination at diagnosis. We adapted Ogata score to MF context including 2 parameters: absolute CD34+ cells count (/µL) and granulocytes to lymphocytes SSC ratio. A score of 1 was attributed to above-threshold values of each parameter. Accordingly, patients were categorized as MFClow (score = 0, 62.0%), MFCint (score = 1, 29.5%), and MFChigh (score = 2, 8.5%). MFClow had significantly longer median OS (not reached) compared to MFCint (55 months) and MFChigh (19 months). We integrated MFC into established models as a substitute of morphological PB blasts count. Patients were reclassified according to MFC-enhanced scores, and concordance (C-) indexes were compared. As regards IPSS, C-indexes were 0.67 and 0.74 for standard and MFC-enhanced model, respectively (Z score - 3.82; p = 0.0001). MFC-enhanced MIPSS70+ model in PMF patients yielded a C-index of 0.78, outperforming its standard counterpart (C-index 0.73; Z score - 2.88, p = 0.004). Our data suggest that the incorporation of MFC-derived parameters, easily attainable from standard assay used for CD34+ cells determination, might help to refine the current prognostic stratification models in myelofibrosis.


Asunto(s)
Mielofibrosis Primaria , Antígenos CD34 , Citometría de Flujo , Humanos , Mutación , Mielofibrosis Primaria/diagnóstico , Mielofibrosis Primaria/genética , Pronóstico
5.
Br J Haematol ; 189(2): 291-302, 2020 04.
Artículo en Inglés | MEDLINE | ID: mdl-31945802

RESUMEN

Survival prediction in essential thrombocythaemia (ET) and polycythaemia vera (PV) is currently based on clinically-derived variables; we examined the possibility of integrating genetic information for predicting survival. To this end, 906 molecularly-annotated patients (416 Mayo Clinic; 490 University of Florence, Italy), including 502 ET and 404 PV, were recruited. Multivariable analysis identified spliceosome mutations to adversely affect overall (SF3B1, SRSF2 in ET and SRSF2 in PV) and myelofibrosis-free (U2AF1, SF3B1 in ET) survival; TP53 mutations predicted leukaemic transformation in ET; "adverse" mutations occurred in 51 (10%) ET and 8 (2%) PV patients. We confirmed the independent survival effect of adverse mutations [hazard ratio (HR) 2·4, 95% CI 1·6-3·5], age >60 years (6·6, 4·6-9·7), male sex (1·8, 1·3-2·4) and leukocytosis ≥11 × 109 /l (1·6, 1·1-2·2), in ET, and adverse mutations (7·8, 3·1-17·0), age >67 years (5·4, 3·6-8·1), leukocytosis ≥15 × 109 /l (2·8, 1·8-4·2) and thrombosis history (2·0, 1·4-2·9), in PV. HR-based risk point allocation allowed development of three-tiered mutation-enhanced international prognostic systems (MIPSS) which were validated in both cohorts and performance was shown to be superior to conventional scoring systems. Spliceosome mutations enhance survival prediction in ET and PV and identify patients at risk for fibrotic progression. TP53 mutations predict leukaemic transformation in ET.


Asunto(s)
Policitemia Vera/genética , Trombocitemia Esencial/genética , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Pronóstico
7.
Curr Oncol Rep ; 21(11): 101, 2019 11 14.
Artículo en Inglés | MEDLINE | ID: mdl-31728739

RESUMEN

PURPOSE OF REVIEW: Chronic myelomonocytic leukemia (CMML) is a clonal hematological malignancy characterized by both dysplastic and proliferative features, with an inherent risk for leukemic transformation. With the help of this review, we aim to summarize key concepts with regards to CMML biology, diagnosis, risk stratification, and therapeutics. RECENT FINDINGS: Based on recent studies, CMML is hallmarked by a relatively low genetic complexity, which contrasts with a compelling phenotypical heterogeneity, largely driven by epigenetic mechanisms. Recent advances in the characterization of CMML biology has led to an improvement in risk-stratification, by means of incorporating prognostically relevant gene mutations. This, however, has not significantly impacted available therapies and outcomes continue to remain poor. Advances in CMML biology have better explained the phenotypic heterogeneity, while continuing to define the genetic and epigenetic landscape. In spite of recent advances, limited effective therapies exist and developing rationally derived therapeutic approaches is much needed.


Asunto(s)
Leucemia Mielomonocítica Crónica/genética , Leucemia Mielomonocítica Crónica/terapia , Cariotipo Anormal , Antineoplásicos/uso terapéutico , Epigénesis Genética , Trasplante de Células Madre Hematopoyéticas , Humanos , Leucemia Mielomonocítica Crónica/diagnóstico , Leucemia Mielomonocítica Crónica/patología , Terapia Molecular Dirigida , Mutación , Pronóstico
9.
Am J Hematol ; 92(11): 1193-1197, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28795425

RESUMEN

Based on suggestive information from recent epidemiologic data and earlier retrospective studies, we revisited the effect of gender on survival in 1,494 patients with essential thrombocythemia (ET). The primary study population included 904 patients from the Mayo Clinic (median age 58 years; 65% females); risk distribution, according to the international prognostic score for ET (IPSET), was 23% high, 42% intermediate and 35% low. Multivariable analysis that included IPSET-relevant risk factors identified male sex (HR 1.6, 95% CI 1.3-2.0), age ≥60 years (HR 4.3, 95% CI 3.4-5.4) and leukocyte count ≥11 × 10(9)/L (HR 1.5, 95% CI 1.3-1.9) as independent predictors of shortened survival. These findings were confirmed by analysis of a separate cohort of 590 ET patients (65% females) from the University of Florence, Italy, with corresponding HRs (95% CI) of 1.6 (1.1-2.5), 4.6 (2.2-9.5) and 1.8 (1.1-2.8). The independent prognostic effect of gender was further corroborated by a separate multivariable analysis against IPSET risk categories; HR (95% CI) for the Mayo Clinic/Florence cohorts were 1.5/1.6 (1.2/1.1-1.8/2.5) for male sex, 6.8/7.5 (5.0/3.1-9.3/18.3) for IPSET high risk and 2.8/4.1 (2.1/1.8-3.8/9.5) for IPSET intermediate risk. Furthermore, the survival disadvantage in men was most apparent in IPSET high risk category and in patients older than 60 years. In both patient cohorts, thrombosis history garnered significance in univariate, but not in multivariable analysis. The observations from the current study suggest that women with ET live longer than their male counterparts and that gender might supersede thrombosis history as a risk variable for overall survival.


Asunto(s)
Trombocitemia Esencial/mortalidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Femenino , Estudios de Seguimiento , Humanos , Masculino , Persona de Mediana Edad , Vigilancia de la Población , Pronóstico , Factores de Riesgo , Factores Sexuales , Análisis de Supervivencia , Trombocitemia Esencial/diagnóstico , Trombocitemia Esencial/epidemiología , Trombocitemia Esencial/etiología , Adulto Joven
10.
Eur J Clin Invest ; 46(8): 683-9, 2016 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-27271054

RESUMEN

BACKGROUND: True essential thrombocythemia (ET) may carry one of the known driver mutations (JAK2, MPL and CALR) or none of them [in triple-negative (3NEG) cases]. The patients' mutational status seems to delineate the clinical manifestations of ET. MATERIALS AND METHODS: We report the data of 183 patients diagnosed with ET strictly according to the WHO 2008 criteria and with a full molecular diagnosis, including the following: 114 patients (62·3%) with JAK2V617F; 25 (13·7%) with CALR type 1 and 19 (10·4%) with CALR type 2; 3 (1·6%) with MPL; 22 (12%) who were 3NEG. Thrombotic risk was assessed by means of the IPSET-thrombosis score (IPSET-T). RESULTS: CALR and 3NEG patients had lower haemoglobin levels and leucocyte count than JAK2 patients. CALR patients, and those with type 2 in particular, had higher mean platelet counts and had extreme thrombocytosis more often than any of the other groups. Based on their IPSET-T stratification, 3NEG- and CALR-mutated patients belonged more frequently to the low-risk group and had a significant more favourable thrombosis-free survival rate than those with JAK2 mutation. CONCLUSION: These findings indicate that the three different molecular markers have a significant impact on the clinical course of true ET, giving rise to different phenotypes of the same disease.


Asunto(s)
Mutación/genética , Trombocitemia Esencial/genética , Trombosis/genética , Adulto , Anciano , Anciano de 80 o más Años , Calreticulina/genética , Femenino , Marcadores Genéticos/genética , Humanos , Janus Quinasa 2/genética , Recuento de Leucocitos , Masculino , Persona de Mediana Edad , Fenotipo , Recuento de Plaquetas , Receptores de Trombopoyetina/genética , Estudios Retrospectivos , Factores de Riesgo
14.
Am J Hematol ; 95(2): E31-E34, 2020 02.
Artículo en Inglés | MEDLINE | ID: mdl-31705546
17.
Bioengineering (Basel) ; 10(5)2023 May 13.
Artículo en Inglés | MEDLINE | ID: mdl-37237661

RESUMEN

Relapsed and/or refractory (R/R) acute myeloid leukemia (AML) is hallmarked by dramatic prognosis. Treatment remains challenging, with allogeneic hematopoietic stem cell transplantation (HSCT) as the only curative option. The BCL-2 inhibitor venetoclax (VEN) has proven to be a promising therapy for AML and is currently the standard of care in combination with hypomethylating agents (HMAs) for newly diagnosed AML patients ineligible for induction chemotherapy. Given its satisfactory safety profile, VEN-based combinations are increasingly being investigated as a part of the therapeutic strategy for R/R AML. The current paper aims to provide a comprehensive review of the main evidence regarding VEN in the setting of R/R AML, with a specific focus on combinational strategies, including HMAs and cytotoxic chemotherapy, as well as different clinical settings, especially in view of the crucial role of HSCT. A discussion of what is known about drug resistance mechanisms and future combinational strategies is also provided. Overall, VEN-based regimes (mainly VEN + HMA) have provided unprecedented salvage treatment opportunities in patients with R/R AML, with low extra-hematological toxicity. On the other hand, the issue of overcoming resistance is one of the most important fields to be addressed in upcoming clinical research.

18.
Cancer Med ; 12(7): 8166-8171, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36708083

RESUMEN

BACKGROUND: Incorporating real-world data in the drug development process allows the improvement of health outcomes by providing better representation of actual patterns of drug safety and efficacy. AIMS AND METHODS: Here, we present the results of a retroprospective, observational real-life study of 154 patients with myelofibrosis treated with ruxolitinib in a real-life setting in seven Italian centers of the MYNERVA project. RESULTS: Median drug exposure was 29 (range, 3-98) months. Discontinuation rate was 27% after a median time of 13 (range, 3-61). While hematological toxicities were in line with previous findings, infections occurred frequently, representing a not negligible cause of discontinuation and death. Anemia, symptoms, and spleen responses were obtained at any time in 23%, 91%, and 68% of patients, respectively; most patients achieved their responses by week 24. Larger splenomegaly and delayed treatment initiation correlated with lower spleen response at 24 weeks. Spleen response was associated with a superior overall survival, regardless of DIPSS. Of interest, both achievement and loss of spleen response had prognostic implications. DISCUSSION AND CONCLUSION: Overall, our findings provide insights on the efficacy and safety of ruxolitinib in a real-world, multicenter cohort of Italian MF patients.


Asunto(s)
Mielofibrosis Primaria , Humanos , Mielofibrosis Primaria/complicaciones , Nitrilos/uso terapéutico , Pirimidinas/uso terapéutico , Italia , Resultado del Tratamiento
19.
Am J Hematol ; 92(8): 833-838, 2017 08.
Artículo en Inglés | MEDLINE | ID: mdl-28457008
20.
Expert Opin Drug Saf ; 20(2): 139-154, 2021 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-33327810

RESUMEN

INTRODUCTION: During the last decade, the development of small molecule inhibitors of Janus kinases (JAKi) contributed to revolutionize the therapeutic landscape of myelofibrosis (MF). JAKi proved to be effective in controlling disease-related symptoms and splenomegaly with remarkable inter-drug variability. However, in some cases the border between clinical efficacy of JAKi and dose-dependent toxicities is narrow leading to sub-optimal dose modifications and/or treatment discontinuation. AREAS COVERED: In the current review, the authors aimed at providing a comprehensive review of the safety profile of JAKi that are currently approved or in advanced clinical development. Also, a short discussion of promising JAKi in early clinical evaluation and molecules 'lost' early in clinical development is provided. Finally, we discuss the possible strategies aimed at strengthening the safety of JAKi while improving the therapeutic efficacy. EXPERT OPINION: Overall, JAKi display a satisfactory risk-benefit ratio, with main toxicities being gastrointestinal or related to the myelo/immunosuppressive effects, generally mild and easily manageable. However, JAKi may be associated with potentially life-threatening toxicities, such as neurological and infectious events. Thus, many efforts are needed in order to optimize JAKi-based therapeutic strategies without burdening patient safety. This could be attempted through drug combinations or the development of more selective molecules.


Asunto(s)
Inhibidores de las Cinasas Janus/administración & dosificación , Mielofibrosis Primaria/tratamiento farmacológico , Relación Dosis-Respuesta a Droga , Desarrollo de Medicamentos , Humanos , Inhibidores de las Cinasas Janus/efectos adversos , Mielofibrosis Primaria/fisiopatología , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA