Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 116
Filtrar
1.
J Alzheimers Dis ; 99(4): 1317-1331, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38788066

RESUMEN

Background: Emerging diagnostic modalities suggest that miRNA profiles within extracellular vesicles (EVs) isolated from peripheral blood specimens may provide a non-invasive diagnostic alternative for dementia and neurodegenerative disorders. Given that EVs confer a protective environment against miRNA enzymatic degradation, the miRNAs enriched in the EV fraction of blood samples could serve as more stable and clinically relevant biomarkers compared to those obtained from serum. Objective: To compare miRNAs isolated from EVs versus serum in blood taken from Alzheimer's disease (AD) dementia patients and control cohorts. Methods: We compared 25 AD patients to 34 individuals who exhibited no cognitive impairments (NCI). Subjects were Singapore residents with Chinese heritage. miRNAs purified from serum versus blood-derived EVs were analyzed for associations with AD dementia and medial temporal atrophy detected by magnetic resonance imaging. Results: Compared to serum-miRNAs, we identified almost twice as many EV-miRNAs associated with AD dementia, and they also correlated more significantly with medial temporal atrophy, a neuroimaging marker of AD-brain pathology. We further developed combination panels of serum-miRNAs and EV-miRNAs with improved performance in identifying AD dementia. Dominant in both panels was miRNA-1290. Conclusions: This data indicates that miRNA profiling from EVs offers diagnostic superiority. This underscores the role of EVs as vectors harboring prognostic biomarkers for neurodegenerative disorders and suggests their potential in yielding novel biomarkers for AD diagnosis.


Asunto(s)
Enfermedad de Alzheimer , Atrofia , Biomarcadores , Vesículas Extracelulares , MicroARNs , Lóbulo Temporal , Humanos , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/diagnóstico , Enfermedad de Alzheimer/sangre , Vesículas Extracelulares/metabolismo , Vesículas Extracelulares/genética , MicroARNs/sangre , MicroARNs/genética , Masculino , Femenino , Anciano , Biomarcadores/sangre , Lóbulo Temporal/patología , Lóbulo Temporal/diagnóstico por imagen , Imagen por Resonancia Magnética , Persona de Mediana Edad , Anciano de 80 o más Años
4.
Cell Rep ; 42(8): 113016, 2023 08 29.
Artículo en Inglés | MEDLINE | ID: mdl-37597186

RESUMEN

Small cell lung cancers (SCLCs) rapidly resist cytotoxic chemotherapy and immune checkpoint inhibitor (ICI) treatments. New, non-cross-resistant therapies are thus needed. SCLC cells are committed into neuroendocrine lineage then maturation arrested. Implicating DNA methyltransferase 1 (DNMT1) in the maturation arrests, we find (1) the repression mark methylated CpG, written by DNMT1, is retained at suppressed neuroendocrine-lineage genes, even as other repression marks are erased; (2) DNMT1 is recurrently amplified, whereas Ten-Eleven-Translocation 2 (TET2), which functionally opposes DNMT1, is deleted; (3) DNMT1 is recruited into neuroendocrine-lineage master transcription factor (ASCL1, NEUROD1) hubs in SCLC cells; and (4) DNMT1 knockdown activated ASCL1-target genes and released SCLC cell-cycling exits by terminal lineage maturation, which are cycling exits that do not require the p53/apoptosis pathway used by cytotoxic chemotherapy. Inhibiting DNMT1/corepressors with clinical compounds accordingly extended survival of mice with chemorefractory and ICI-refractory, p53-null, disseminated SCLC. Lineage commitment of SCLC cells can hence be leveraged into non-cytotoxic therapy able to treat chemo/ICI-refractory SCLC.


Asunto(s)
Neoplasias Pulmonares , Carcinoma Pulmonar de Células Pequeñas , Animales , Ratones , Proteína p53 Supresora de Tumor/genética , Carcinoma Pulmonar de Células Pequeñas/tratamiento farmacológico , Ciclo Celular , División Celular , Neoplasias Pulmonares/tratamiento farmacológico
5.
Diabetes Spectr ; 36(2): 161-170, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37193209

RESUMEN

Objective: To assess whether an electronic health record (EHR)-based diabetes intensification tool can improve the rate of A1C goal attainment among patients with type 2 diabetes and an A1C ≥8%. Methods: An EHR-based tool was developed and sequentially implemented in a large, integrated health system using a four-phase, stepped-wedge design (single pilot site [phase 1] and then three practice site clusters [phases 2-4]; 3 months/phase), with full implementation during phase 4. A1C outcomes, tool usage, and treatment intensification metrics were compared retrospectively at implementation (IMP) sites versus nonimplementation (non-IMP) sites with sites matched on patient population characteristics using overlap propensity score weighting. Results: Overall, tool utilization was low among patient encounters at IMP sites (1,122 of 11,549 [9.7%]). During phases 1-3, the proportions of patients achieving the A1C goal (<8%) were not significantly improved between IMP and non-IMP sites at 6 months (range 42.9-46.5%) or 12 months (range 46.5-53.1%). In phase 3, fewer patients at IMP sites versus non-IMP sites achieved the goal at 12 months (46.7 vs. 52.3%, P = 0.02). In phases 1-3, mean changes in A1C from baseline to 6 and 12 months (range -0.88 to -1.08%) were not significantly different between IMP and non-IMP sites. Times to intensification were similar between IMP and non-IMP sites. Conclusion: Utilization of a diabetes intensification tool was low and did not influence rates of A1C goal attainment or time to treatment intensification. The low level of tool adoption is itself an important finding highlighting the problem of therapeutic inertia in clinical practice. Testing additional strategies to better incorporate, increase acceptance of, and improve proficiency with EHR-based intensification tools is warranted.

6.
NPJ Digit Med ; 5(1): 106, 2022 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-35896817

RESUMEN

Deep learning (DL) from electronic health records holds promise for disease prediction, but systematic methods for learning from simulated longitudinal clinical measurements have yet to be reported. We compared nine DL frameworks using simulated body mass index (BMI), glucose, and systolic blood pressure trajectories, independently isolated shape and magnitude changes, and evaluated model performance across various parameters (e.g., irregularity, missingness). Overall, discrimination based on variation in shape was more challenging than magnitude. Time-series forest-convolutional neural networks (TSF-CNN) and Gramian angular field(GAF)-CNN outperformed other approaches (P < 0.05) with overall area-under-the-curve (AUCs) of 0.93 for both models, and 0.92 and 0.89 for variation in magnitude and shape with up to 50% missing data. Furthermore, in a real-world assessment, the TSF-CNN model predicted T2D with AUCs reaching 0.72 using only BMI trajectories. In conclusion, we performed an extensive evaluation of DL approaches and identified robust modeling frameworks for disease prediction based on longitudinal clinical measurements.

7.
Clin Cancer Res ; 28(21): 4689-4701, 2022 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-35776130

RESUMEN

PURPOSE: Advanced/metastatic forms of clear-cell renal cell carcinomas (ccRCC) have limited therapeutic options. Genome-wide genetic screens have identified cellular dependencies in many cancers. Using the Broad Institute/Novartis combined short hairpin RNA (shRNA) dataset, and cross-validation with the CRISPR/Cas9 DepMap (21Q3) dataset, we sought therapeutically actionable dependencies in kidney lineage cancers. EXPERIMENTAL DESIGN: We identified preferential genetic dependencies in kidney cancer cells versus other lineages. BCL2L1, which encodes the BCL-XL antiapoptotic protein, scored as the top actionable dependency. We validated this finding using genetic and pharmacologic tools in a panel of ccRCC cell lines. Select BCL-XL-dependent (versus independent) cell lines were then transcriptionally profiled to identify biomarkers and mechanistic drivers of BCL-XL dependence. Cell-based studies (in vitro and in vivo) and clinical validations were used to address physiologic relevance. RESULTS: Inactivation of BCL-XL, but not BCL-2, led to fitness defects in renal cancer cells, and sensitized them to chemotherapeutics. Transcriptomic profiling identified a "BCL-XL dependency" signature, including an elevated mesenchymal gene signature. A mesenchymal state was both necessary and sufficient to confer increased BCL-XL dependence. The "BCL-XL dependency" signature was observed in approximately 30% of human ccRCCs, which were also associated with worse clinical outcomes. Finally, an orally bioavailable BCL-XL inhibitor, A-1331852, showed antitumor efficacy in vivo. CONCLUSIONS: Our studies uncovered an unexpected link between cell state and BCL-XL dependence in ccRCC. Therapeutic agents that specifically target BCL-XL are available. Our work justifies testing the utility of BCL-XL blockade to target, likely, a clinically aggressive subset of human kidney cancers. See related commentary by Wang et al., p. 4600.


Asunto(s)
Carcinoma de Células Renales , Neoplasias Renales , Humanos , Carcinoma de Células Renales/tratamiento farmacológico , Carcinoma de Células Renales/genética , Línea Celular Tumoral , Proteína bcl-X/genética , Proteína bcl-X/metabolismo , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Apoptosis/genética , Neoplasias Renales/tratamiento farmacológico , Neoplasias Renales/genética , ARN Interferente Pequeño
8.
EBioMedicine ; 80: 104059, 2022 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-35617825

RESUMEN

BACKGROUND: Haploinsufficiency (HI) resulting from deletion of the long arm of chromosome 5 [del(5q)] and the accompanied loss of heterozygosity are likely key pathogenic factors in del(5q) myeloid neoplasia (MN) although the consequences of del(5q) have not been yet clarified. METHODS: Here, we explored mutations, gene expression and clinical phenotypes of 388 del(5q) vs. 841 diploid cases with MN [82% myelodysplastic syndromes (MDS)]. FINDINGS: Del(5q) resulted as founder (better prognosis) or secondary hit (preceded by TP53 mutations). Using Bayesian prediction analyses on 57 HI marker genes we established the minimal del(5q) gene signature that distinguishes del(5q) from diploid cases. Clusters of diploid cases mimicking the del(5q) signature support the overall importance of del(5q) genes in the pathogenesis of MDS in general. Sub-clusters within del(5q) patients pointed towards the inherent intrapatient heterogeneity of HI genes. INTERPRETATION: The underlying clonal expansion drive results from a balance between the "HI-driver" genes (e.g., CSNK1A1, CTNNA1, TCERG1) and the proapoptotic "HI-anti-drivers" (e.g., RPS14, PURA, SIL1). The residual essential clonal expansion drive allows for selection of accelerator mutations such as TP53 (denominating poor) and CSNK1A1 mutations (with a better prognosis) which overcome pro-apoptotic genes (e.g., p21, BAD, BAX), resulting in a clonal expansion. In summary, we describe the complete picture of del(5q) MN identifying the crucial genes, gene clusters and clonal hierarchy dictating the clinical course of del(5q) patients. FUNDING: Torsten Haferlach Leukemia Diagnostics Foundation. US National Institute of Health (NIH) grants R35 HL135795, R01HL123904, R01 HL118281, R01 HL128425, R01 HL132071, and a grant from Edward P. Evans Foundation.


Asunto(s)
Deleción Cromosómica , Síndromes Mielodisplásicos , Teorema de Bayes , Cromosomas Humanos Par 5 , Factores de Intercambio de Guanina Nucleótido/genética , Haploinsuficiencia/genética , Humanos , Mutación , Síndromes Mielodisplásicos/diagnóstico , Síndromes Mielodisplásicos/genética , Factores de Elongación Transcripcional/genética
9.
J Pediatr Hematol Oncol ; 44(7): e1006-e1015, 2022 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-35082244

RESUMEN

Langerhans cell histiocytosis (LCH) is a disorder with highly diverse clinical manifestations. We explored if age, sex, race, organ system involved, and therapy approaches determine patient survival in the era of modern treatments. LCH patient data reported to the Surveillance, Epidemiology, and End Results (SEER) program in 2010-2016 (n=1282; age: 0 to 100 y) was analyzed. Age-specific LCH incidence flattening to a low level suggests an age cutoff for pediatric patients of 20 years. The overall survival probability is lower for patients 21 to 100 years old ( P <0.0001), irrespective of sex and race. The commonest sites involved in the 0- to 20-year age group were bone, skin, and bone marrow; this shifted to lung, bone, and skin as the commonest disease sites in patients 21 to 100 years of age. The treatments applied differed between age groups, as younger versus older patients were more likely to receive chemotherapy-based treatment (48.4% vs. 17%; P <0.0001). There also was a trend toward nonwhite versus white patients being less likely to receive chemotherapy-based treatment (31.7% vs. 38.2%; P =0.067). Whereas there are treatment disparities related to LCH patient age and perhaps race, patient age is the strongest predictor of survival, with patients 21 to 100 years of age with lung, lymph node, skin, and bone marrow disease having the worst outcomes ( P <0.0001).


Asunto(s)
Enfermedades de la Médula Ósea , Histiocitosis de Células de Langerhans , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Huesos/patología , Niño , Preescolar , Histiocitosis de Células de Langerhans/epidemiología , Histiocitosis de Células de Langerhans/patología , Histiocitosis de Células de Langerhans/terapia , Humanos , Incidencia , Lactante , Recién Nacido , Ganglios Linfáticos/patología , Persona de Mediana Edad , Estudios Retrospectivos , Estados Unidos/epidemiología , Adulto Joven
10.
Leuk Lymphoma ; 63(2): 335-343, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34521300

RESUMEN

Using the National Cancer Database, we identified 10,290 patients with newly diagnosed mantle cell lymphoma (MCL) treated with chemotherapy with or without upfront autologous stem cell transplantation (ASCT). Only 17% of patients underwent ASCT. Patients who underwent ASCT were younger and more likely to have lower comorbidity scores, private insurance, higher income and education, and treatment received at an academic facility. On multivariable analysis, age, comorbidity index, insurance type, the transition of care, facility type, distance to facility, and diagnosis year were predictive for ASCT use. ASCT use was associated with improved 5-year overall survival in younger (82% vs. 64%, p < .001) and older (70% vs. 40%, p < .001) patients, which was retained in the matched propensity score and 12-month analyses. Female gender, the diagnosis year ≥2009, private insurance, higher income, and education were associated with superior survival, whereas Black race and higher comorbidities predicted inferior survival.


Asunto(s)
Trasplante de Células Madre Hematopoyéticas , Linfoma de Células del Manto , Adulto , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Femenino , Humanos , Linfoma de Células del Manto/tratamiento farmacológico , Linfoma de Células del Manto/terapia , Factores Socioeconómicos , Trasplante de Células Madre , Trasplante Autólogo
11.
Cell Rep ; 36(12): 109747, 2021 09 21.
Artículo en Inglés | MEDLINE | ID: mdl-34551289

RESUMEN

PBRM1, a subunit of the PBAF coactivator complex that transcription factors use to activate target genes, is genetically inactivated in almost all clear cell renal cell cancers (RCCs). Using unbiased proteomic analyses, we find that PAX8, a master transcription factor driver of proximal tubule epithelial fates, recruits PBRM1/PBAF. Reverse analyses of the PAX8 interactome confirm recruitment specifically of PBRM1/PBAF and not functionally similar BAF. More conspicuous in the PAX8 hub in RCC cells, however, are corepressors, which functionally oppose coactivators. Accordingly, key PAX8 target genes are repressed in RCC versus normal kidneys, with the loss of histone lysine-27 acetylation, but intact lysine-4 trimethylation, activation marks. Re-introduction of PBRM1, or depletion of opposing corepressors using siRNA or drugs, redress coregulator imbalance and release RCC cells to terminal epithelial fates. These mechanisms thus explain RCC resemblance to the proximal tubule lineage but with suppression of the late-epithelial program that normally terminates lineage-precursor proliferation.


Asunto(s)
Carcinoma de Células Renales/patología , Diferenciación Celular , Proteínas de Unión al ADN/metabolismo , Túbulos Renales Proximales/metabolismo , Factor de Transcripción PAX8/metabolismo , Factores de Transcripción/metabolismo , Animales , Carcinoma de Células Renales/metabolismo , Línea Celular Tumoral , Proliferación Celular , Proteínas de Unión al ADN/antagonistas & inhibidores , Proteínas de Unión al ADN/genética , Histonas/metabolismo , Humanos , Neoplasias Renales/metabolismo , Neoplasias Renales/patología , Túbulos Renales Proximales/citología , Masculino , Ratones , Ratones Desnudos , Mutagénesis , Factor de Transcripción PAX8/genética , Mapas de Interacción de Proteínas , Interferencia de ARN , ARN Interferente Pequeño/metabolismo , Factores de Transcripción/antagonistas & inhibidores , Factores de Transcripción/genética , Activación Transcripcional , Trasplante Heterólogo
12.
Blood ; 138(19): 1885-1895, 2021 11 11.
Artículo en Inglés | MEDLINE | ID: mdl-34075412

RESUMEN

Although genomic alterations drive the pathogenesis of acute myeloid leukemia (AML), traditional classifications are largely based on morphology, and prototypic genetic founder lesions define only a small proportion of AML patients. The historical subdivision of primary/de novo AML and secondary AML has shown to variably correlate with genetic patterns. The combinatorial complexity and heterogeneity of AML genomic architecture may have thus far precluded genomic-based subclassification to identify distinct molecularly defined subtypes more reflective of shared pathogenesis. We integrated cytogenetic and gene sequencing data from a multicenter cohort of 6788 AML patients that were analyzed using standard and machine learning methods to generate a novel AML molecular subclassification with biologic correlates corresponding to underlying pathogenesis. Standard supervised analyses resulted in modest cross-validation accuracy when attempting to use molecular patterns to predict traditional pathomorphologic AML classifications. We performed unsupervised analysis by applying the Bayesian latent class method that identified 4 unique genomic clusters of distinct prognoses. Invariant genomic features driving each cluster were extracted and resulted in 97% cross-validation accuracy when used for genomic subclassification. Subclasses of AML defined by molecular signatures overlapped current pathomorphologic and clinically defined AML subtypes. We internally and externally validated our results and share an open-access molecular classification scheme for AML patients. Although the heterogeneity inherent in the genomic changes across nearly 7000 AML patients was too vast for traditional prediction methods, machine learning methods allowed for the definition of novel genomic AML subclasses, indicating that traditional pathomorphologic definitions may be less reflective of overlapping pathogenesis.


Asunto(s)
Leucemia Mieloide Aguda/genética , Aprendizaje Automático , Teorema de Bayes , Citogenética , Regulación Leucémica de la Expresión Génica , Genómica , Humanos , Leucemia Mieloide Aguda/clasificación , Leucemia Mieloide Aguda/diagnóstico , Mutación , Neoplasias Primarias Secundarias/clasificación , Neoplasias Primarias Secundarias/diagnóstico , Neoplasias Primarias Secundarias/genética , Translocación Genética
13.
PLoS One ; 16(5): e0250939, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34003826

RESUMEN

PURPOSE: The long-term survival of uveal melanoma patients in the US is not known. We compared long-term survival estimates using relative survival, excess absolute risk (EAR), Kaplan-Meier (KM), and competing risk analyses. SETTING: Population based cohort study. STUDY POPULATION: Pooled databases from Surveillance, Epidemiology, and End Results data (SEER, SEER-9+SEER-13+SEER-18). MAIN OUTCOME MEASURE: Overall Survival (OS), Metastasis Free Survival (MFS) and relative survival, computed directly or estimated via a model fitted to excess mortality. RESULTS: There were 10678 cases of uveal melanoma spanning a period of 42 years (1975-2016). The median age at diagnosis was 63 years (range 3-99). Over half the patients were still alive at the end of 2016 (53%, 5625). The KM estimates of MFS were 0.729 (0.719, 0.74), 0.648 (0.633, 0.663), and 0.616 (0.596, 0.636) at 10, 20, and 30 years, respectively. The cumulative probabilities of melanoma metastatic death at 10, 20 and 30 years were 0.241 (0.236, 0.245), 0.289 (0.283, 0.294), and 0.301 (0.295, 0.307). In the first 5 years since diagnosis of uveal melanoma, the proportion of deaths attributable to uveal melanoma were 1.3 with rapid fall after 10 years. Death due to melanoma were rare beyond 20 years. Relative survival (RS) plateaued to ~60% across 20 to 30 years. EAR parametric modeling yielded a survival probability of 57%. CONCLUSIONS: Relative survival methods can be used to estimate long term survival of uveal melanoma patients without knowing the exact cause of death. RS and EAR provide more realistic estimates as they compare the survival to that of a normal matched population. Death due to melanoma were rare beyond 20 years with normal life expectancy reached at 25 years after primary therapy.


Asunto(s)
Melanoma/mortalidad , Neoplasias de la Úvea/mortalidad , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Causas de Muerte , Niño , Preescolar , Estudios de Cohortes , Bases de Datos Factuales , Femenino , Humanos , Masculino , Melanoma/epidemiología , Melanoma/patología , Persona de Mediana Edad , Medición de Riesgo/métodos , Programa de VERF , Tasa de Supervivencia , Estados Unidos/epidemiología , Neoplasias de la Úvea/epidemiología , Neoplasias de la Úvea/patología , Adulto Joven
14.
NPJ Breast Cancer ; 7(1): 21, 2021 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-33654083

RESUMEN

The increased incidence of secondary hematologic malignancies (SHM) is a well-known, potentially fatal, complication after cancer treatment. It is unknown if patients with ductal carcinoma in situ (DCIS) of the breast treated with external beam radiotherapy (RT) and who survive long-term have increased risks of secondary hematologic malignancies (SHM), especially for low/intermediate-risk subsets with limited benefits from RT. DCIS patients in Surveillance, Epidemiology, and End Results (SEER) registries (1975-2016) were identified. Relative risks (RR), hazard ratio (HR), and standardized incidence ratios (SIR) were calculated to assess the SHM risk and subsequent survival times. SHM development, defined as a nonsynchronous SHM occurring ≥1 year after DCIS diagnosis, was our primary endpoint. Of 184,363 eligible patients with DCIS, 77,927 (42.3%) in the RT group, and 106,436 (57.7%) in the non-RT group, 1289 developed SHMs a median of 6.4 years (interquartile range, 3.5 to 10.3 years) after their DCIS diagnosis. Compared with DCIS patients in the non-RT group, RT was associated with increased early risk of developing acute lymphoblastic leukemia (ALL; hazard ratio, 3.15; 95% CI, 1.21 to 8.17; P = 0.02), and a delayed risk of non-Hodgkin lymphoma (NHL; hazard ratio, 1.33; 95% CI, 1.09 to 1.62; P < 0.001). This increased risk of ALL and NHL after RT was also observed in subgroup analyses restricted to low/intermediate-risk DCIS. In summary, our data suggest that RT after breast conserving surgery for DCIS patients should be cautiously tailored, especially for low and intermediate-risk patients. Long-term SHM surveillance after DCIS diagnosis is warranted.

15.
Blood Cancer Discov ; 2(2): 146-161, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33681816

RESUMEN

TET2 is frequently mutated in myeloid neoplasms. Genetic TET2 deficiency leads to skewed myeloid differentiation and clonal expansion, but minimal residual TET activity is critical for survival of neoplastic progenitor and stem cells. Consistent with mutual exclusivity of TET2 and neomorphic IDH1/2 mutations, here we report that IDH1/2 mutant-derived 2-hydroxyglutarate is synthetically lethal to TET-dioxygenase deficient cells. In addition, a TET-selective small molecule inhibitor decreased cytosine hydroxymethylation and restricted clonal outgrowth of TET2 mutant, but not normal hematopoietic precursor cells in vitro and in vivo. While TET-inhibitor phenocopied somatic TET2 mutations, its pharmacologic effects on normal stem cells were, unlike mutations, reversible. Treatment with TET inhibitor suppressed the clonal evolution of TET2 mutant cells in murine models and TET2-mutated human leukemia xenografts. These results suggest that TET inhibitors may constitute a new class of targeted agents in TET2 mutant neoplasia.


Asunto(s)
Dioxigenasas , Leucemia , Animales , Proteínas de Unión al ADN/genética , Hematopoyesis/genética , Humanos , Ratones , Proteínas Proto-Oncogénicas/genética
16.
Clin Cancer Res ; 27(7): 2038-2049, 2021 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-33542075

RESUMEN

PURPOSE: Glioblastoma (GBM) immunotherapy clinical trials are generally initiated after standard-of-care treatment-including surgical resection, perioperative high-dose steroid therapy, chemotherapy, and radiation treatment-has either begun or failed. However, the impact of these interventions on the antitumoral immune response is not well studied. While discoveries regarding the impact of chemotherapy and radiation on immune response have been made and translated into clinical trial design, the impact of surgical resection and steroids on the antitumor immune response has yet to be determined. EXPERIMENTAL DESIGN: We developed a murine model integrating tumor resection and steroid treatment and used flow cytometry to analyze systemic and local immune changes. These mouse model findings were validated in a cohort of 95 patients with primary GBM. RESULTS: Using our murine resection model, we observed a systemic reduction in lymphocytes corresponding to increased tumor volume and decreased circulating lymphocytes that was masked by dexamethasone treatment. The reduction in circulating T cells was due to reduced CCR7 expression, resulting in T-cell sequestration in lymphoid organs and the bone marrow. We confirmed these findings in a cohort of patients with primary GBM and found that prior to steroid treatment, circulating lymphocytes inversely correlated with tumor volume. Finally, we demonstrated that peripheral lymphocyte content varies with progression-free survival and overall survival, independent of tumor volume, steroid use, or molecular profiles. CONCLUSIONS: These data reveal that prior to intervention, increased tumor volume corresponds with reduced systemic immune function and that peripheral lymphocyte counts are prognostic when steroid treatment is taken into account.


Asunto(s)
Neoplasias Encefálicas/inmunología , Glioblastoma/inmunología , Anciano , Animales , Neoplasias Encefálicas/mortalidad , Neoplasias Encefálicas/patología , Neoplasias Encefálicas/terapia , Dexametasona/uso terapéutico , Modelos Animales de Enfermedad , Femenino , Glioblastoma/mortalidad , Glioblastoma/patología , Glioblastoma/terapia , Humanos , Tolerancia Inmunológica , Inmunofenotipificación , Recuento de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Persona de Mediana Edad , Carga Tumoral
17.
Semin Hematol ; 58(1): 35-44, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33509441

RESUMEN

One mechanism by which lymphoid malignancies resist standard apoptosis-intending (cytotoxic) treatments is genetic attenuation of the p53/p16-CDKN2A apoptosis axis. Depletion of the epigenetic protein DNA methyltransferase 1 (DNMT1) using the deoxycytidine analog decitabine is a validated approach to cytoreduce malignancy independent of p53/p16. In vivo decitabine activity, however, is restricted by rapid catabolism by cytidine deaminase (CDA). We, therefore, combined decitabine with the CDA-inhibitor tetrahydrouridine and conducted a pilot clinical trial in patients with relapsed lymphoid malignancies: the doses of tetrahydrouridine/decitabine used (∼10/0.2 mg/kg orally (PO) 2×/week) were selected for the molecular pharmacodynamic objective of non-cytotoxic, S-phase dependent, DNMT1-depletion, guided by previous Phase 1 studies. Patients with relapsed/refractory B- or T-cell malignancies (n = 7) were treated for up to 18 weeks. Neutropenia without concurrent thrombocytopenia is an expected toxicity of DNMT1-depletion and occurred in all patients (Grade 3/4). Subjective and objective clinical improvements occurred in 4 of 7 patients, but these responses were lost upon treatment interruptions and reductions to manage neutropenia. We thus performed parallel experiments in a preclinical in vivo model of lymphoma to identify regimen refinements that might sustain DNMT1-targeting in malignant cells but limit neutropenia. We found that timed-alternation of decitabine with the related molecule 5-azacytidine, and combination with inhibitors of CDA and de novo pyrimidine synthesis could leverage feedback responses of pyrimidine metabolism to substantially increase lymphoma cytoreduction but with less neutropenia. In sum, regimen innovations beyond incorporation of a CDA-inhibitor are needed to sustain decitabine DNMT1-targeting and efficacy against chemo-resistant lymphoid malignancy. Such potential solutions were explored in preclinical in vivo studies.


Asunto(s)
Antimetabolitos Antineoplásicos , Tetrahidrouridina , Antimetabolitos Antineoplásicos/uso terapéutico , Azacitidina/farmacología , Azacitidina/uso terapéutico , Decitabina/farmacología , Decitabina/uso terapéutico , Epigénesis Genética , Humanos , Linfoma/tratamiento farmacológico , Proyectos Piloto , Tetrahidrouridina/farmacología , Tetrahidrouridina/uso terapéutico
18.
JAMA Ophthalmol ; 139(2): 174-181, 2021 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-33355644

RESUMEN

Importance: The extent to which uveal melanoma is cured by ocular therapy is not known. Objective: To estimate cured fractions (CF) of uveal melanoma using combination of institutional and Surveillance, Epidemiology, and End Results (SEER) data. Design, Setting, and Participants: Integrative analysis of 42 years of SEER data (1975-2016) with 25 years (1993-2018) of complementary institutional data. The analysis included SEER US patients and molecularly prognosticated patients in the United States and Europe. Three SEER databases (SEER-9, SEER-13, and SEER-18) were merged. A total of 10 678 SEER cases of uveal melanoma diagnosed from 1975 to 2016 using International Classification of Disease for Oncology morphology codes 8720-8790 (for melanoma) and site codes C69.2-4 (for choroid, ciliary body, and iris) were downloaded April 16, 2019. The institutional data included 5 institutional cohorts of 788 molecularly prognosticated patients (diagnosed prior to July 2019) with 3115 person-years at risk of death and 262 observed deaths. Main Outcomes and Measures: Excess absolute risks of death (EAR) and cured fraction (CF) indicates lifetime area under the EAR curve. These are applied to populations and subpopulations. Results: The SEER EAR, with sexes and races pooled, can be modeled as a sum of 2 waves. The first wave peaks at approximately 3 years and is negligible by 15 years, at which time the second wave peaks. Institutional data suggest that the first wave is owing to BAP1 mutant cases (204 of 355 [57.5%]; 95% CI, 52%-63%) and that the second wave is owing to BAP1 wild-type SF3B1 mutant cases (60 of 355 [17%]; 95% CI, 13%-21%). There is also a third group with a low flat EAR time course (91 of 355 [25.5%]; 95% CI, 21%-30%). The overall statistical CF of 60% is reached by approximately 25 years. Conclusions and Relevance: These findings suggest that the benefits of ocular therapy for curing uveal melanoma may be questionable because statistical cures reflect deaths of poor prognosis cases and survival of good prognosis cases. Changes in uveal melanoma patient management may be needed to improve survival.


Asunto(s)
Melanoma/terapia , Neoplasias de la Úvea/terapia , Adolescente , Adulto , Anciano , Anciano de 80 o más Años , Biomarcadores de Tumor/genética , Femenino , Humanos , Masculino , Melanoma/diagnóstico , Melanoma/genética , Melanoma/mortalidad , Persona de Mediana Edad , Mutación , Fosfoproteínas/genética , Factores de Empalme de ARN/genética , Medición de Riesgo , Factores de Riesgo , Programa de VERF , Factores de Tiempo , Resultado del Tratamiento , Proteínas Supresoras de Tumor/genética , Ubiquitina Tiolesterasa/genética , Estados Unidos/epidemiología , Neoplasias de la Úvea/diagnóstico , Neoplasias de la Úvea/genética , Neoplasias de la Úvea/mortalidad , Adulto Joven
19.
Radiat Environ Biophys ; 60(1): 41-47, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33125593

RESUMEN

A model of post-diagnosis chronic myeloid leukemia (CML) dynamics across treatment cessations is applied here to pre-diagnosis scenarios of A-bomb survivors. The main result is that perturbing two parameters of a two-state simplification of this model captures the essence of two A-bomb survivor mysteries: (1) in those exposed to > 1 Sv in Hiroshima, four of six female onsets arose as a cluster in 1969-1974, well after 5-10-year latencies expected and observed in two of six female- and nine of ten male cases (about one background case was expected in this high-dose cohort); and (2) no Nagasaki adult cases exposed to > 0.2 Sv were observed though about nine were expected (~ 1.5 background + ~ 7.5 radiation-induced). Overall, it is concluded that: (1) whole-body radiation co-creates malignant and benign BCR-ABL clones; (2) benign clones are more likely to act as anti-CML vaccines in females than in males; (3) the Hong Kong flu of 1968 (and H3N2 seasonal flu thereafter) exhausted anti-CML immunity, thereby releasing radiation-induced clones latent in high-dose Hiroshima females; and (4) benign cells of 1-2 are CD4+ as human T-cell leukemia-lymphoma virus-1 endemic to Nagasaki but not Hiroshima expands numbers of such cells. The next goal is to see if these conclusions can be substantiated using banked A-bomb survivor blood samples.


Asunto(s)
Supervivientes a la Bomba Atómica , Leucemia Mielógena Crónica BCR-ABL Positiva/diagnóstico , Leucemia Inducida por Radiación/diagnóstico , Modelos Biológicos , Linfocitos T CD4-Positivos/inmunología , Femenino , Humanos , Subtipo H3N2 del Virus de la Influenza A , Gripe Humana/inmunología , Japón/epidemiología , Leucemia Mielógena Crónica BCR-ABL Positiva/inmunología , Leucemia Inducida por Radiación/inmunología , Masculino , Células Madre Neoplásicas/inmunología
20.
Ophthalmol Retina ; 5(6): 536-542, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-32979556

RESUMEN

PURPOSE: To investigate conditional survival in patients with uveal melanoma in the United States. DESIGN: Cohort study. PARTICIPANTS: Patients were identified using International Classification of Disease for Oncology, Third Edition, codes for both morphologic features (melanoma, 8720-8790) and site (retina, C69.2; choroid, C69.3; and ciliary body, C69.4) from 1975 through 2011 using the Surveillance, Epidemiology, and End Results (SEER) database SEER 18. METHODS: Observed metastasis-free survival (MFS) and overall survival (OS) were estimated using the Kaplan-Meier method. Conditional metastasis-free survival (cMFS) and conditional overall survival were calculated based on the observed MFS and OS. Relative survival also was calculated using the actuarial method. Survival to 5 and 10 years after diagnosis were calculated, conditioned on various numbers of years already survived. MAIN OUTCOME MEASURES: Conditional MFS, conditional OS, and conditional relative survival. RESULTS: A total of 6863 cases of uveal melanoma were identified. Median follow-up among survivors was 11 years. During follow-up, 3883 patients died of any cause, and of these, 2131 deaths were the result of metastatic uveal melanoma. The nonconditional 5-year MFS was 80%. After surviving 1, 2, 3, or 4 years after diagnosis, the 5-year cMFS estimates increased to 82%, 87%, 92%, and 96%, respectively. The nonconditional MFS at 10 years was estimated to be 69%. After having survived 5, 6, 7, 8, or 9 years after diagnosis, the 10-year cMFS estimates increased to 87%, 90%, 93%, 96%, and 98%, respectively. This result pattern was confirmed with estimates of relative survival. CONCLUSIONS: Conditional survival estimates of uveal melanoma improve with time since primary diagnosis. Among patients who already have survived for at least 5 years, 10-year conditional survival rates are high. Conditional survival analysis can provide specific guidance for counselling patients.


Asunto(s)
Predicción , Melanoma/mortalidad , Programa de VERF , Neoplasias de la Úvea/mortalidad , Supervivencia sin Enfermedad , Europa (Continente)/epidemiología , Estudios de Seguimiento , Humanos , Melanoma/diagnóstico , Melanoma/secundario , Metástasis de la Neoplasia , Tasa de Supervivencia/tendencias , Neoplasias de la Úvea/diagnóstico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...