Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Cell ; 160(1-2): 74-87, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25543153

RESUMEN

Type 2 innate lymphoid cells (ILC2s), an innate source of the type 2 cytokines interleukin (IL)-5 and -13, participate in the maintenance of tissue homeostasis. Although type 2 immunity is critically important for mediating metabolic adaptations to environmental cold, the functions of ILC2s in beige or brown fat development are poorly defined. We report here that activation of ILC2s by IL-33 is sufficient to promote the growth of functional beige fat in thermoneutral mice. Mechanistically, ILC2 activation results in the proliferation of bipotential adipocyte precursors (APs) and their subsequent commitment to the beige fat lineage. Loss- and gain-of-function studies reveal that ILC2- and eosinophil-derived type 2 cytokines stimulate signaling via the IL-4Rα in PDGFRα(+) APs to promote beige fat biogenesis. Together, our results highlight a critical role for ILC2s and type 2 cytokines in the regulation of adipocyte precursor numbers and fate, and as a consequence, adipose tissue homeostasis. PAPERCLIP:


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Linfocitos/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proliferación Celular , Femenino , Interleucina-13/metabolismo , Interleucina-33 , Interleucinas/inmunología , Linfocitos/citología , Masculino , Ratones , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Receptores de Interleucina-4/metabolismo , Transducción de Señal , Células Madre/metabolismo
2.
Cell ; 153(2): 376-88, 2013 Apr 11.
Artículo en Inglés | MEDLINE | ID: mdl-23582327

RESUMEN

In vertebrates, activation of innate immunity is an early response to injury, implicating it in the regenerative process. However, the mechanisms by which innate signals might regulate stem cell functionality are unknown. Here, we demonstrate that type 2 innate immunity is required for regeneration of skeletal muscle after injury. Muscle damage results in rapid recruitment of eosinophils, which secrete IL-4 to activate the regenerative actions of muscle resident fibro/adipocyte progenitors (FAPs). In FAPs, IL-4/IL-13 signaling serves as a key switch to control their fate and functions. Activation of IL-4/IL-13 signaling promotes proliferation of FAPs to support myogenesis while inhibiting their differentiation into adipocytes. Surprisingly, type 2 cytokine signaling is also required in FAPs, but not in myeloid cells, for rapid clearance of necrotic debris, a process that is necessary for timely and complete regeneration of tissues.


Asunto(s)
Inmunidad Innata , Desarrollo de Músculos , Músculo Esquelético/citología , Músculo Esquelético/lesiones , Transducción de Señal , Animales , Proteínas Cardiotóxicas de Elápidos , Eosinófilos/fisiología , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-4/genética , Interleucina-4/metabolismo , Ratones , Músculo Esquelético/fisiología , Células Mieloides/metabolismo , Receptores de Superficie Celular/metabolismo , Regeneración , Factor de Transcripción STAT6/metabolismo
3.
Nature ; 480(7375): 104-8, 2011 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-22101429

RESUMEN

All homeotherms use thermogenesis to maintain their core body temperature, ensuring that cellular functions and physiological processes can continue in cold environments. In the prevailing model of thermogenesis, when the hypothalamus senses cold temperatures it triggers sympathetic discharge, resulting in the release of noradrenaline in brown adipose tissue and white adipose tissue. Acting via the ß(3)-adrenergic receptors, noradrenaline induces lipolysis in white adipocytes, whereas it stimulates the expression of thermogenic genes, such as PPAR-γ coactivator 1a (Ppargc1a), uncoupling protein 1 (Ucp1) and acyl-CoA synthetase long-chain family member 1 (Acsl1), in brown adipocytes. However, the precise nature of all the cell types involved in this efferent loop is not well established. Here we report in mice an unexpected requirement for the interleukin-4 (IL-4)-stimulated program of alternative macrophage activation in adaptive thermogenesis. Exposure to cold temperature rapidly promoted alternative activation of adipose tissue macrophages, which secrete catecholamines to induce thermogenic gene expression in brown adipose tissue and lipolysis in white adipose tissue. Absence of alternatively activated macrophages impaired metabolic adaptations to cold, whereas administration of IL-4 increased thermogenic gene expression, fatty acid mobilization and energy expenditure, all in a macrophage-dependent manner. Thus, we have discovered a role for alternatively activated macrophages in the orchestration of an important mammalian stress response, the response to cold.


Asunto(s)
Catecolaminas/metabolismo , Activación de Macrófagos , Macrófagos/fisiología , Estrés Fisiológico/fisiología , Termogénesis/fisiología , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Temperatura Corporal/genética , Células Cultivadas , Frío , Metabolismo Energético , Regulación de la Expresión Génica , Humanos , Interleucina-4 , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Células U937
4.
Proc Natl Acad Sci U S A ; 110(24): 9914-9, 2013 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-23716700

RESUMEN

The liver is a central organ for the synthesis and storage of nutrients, production of serum proteins and hormones, and breakdown of toxins and metabolites. Because the liver is susceptible to toxin- or pathogen-mediated injury, it maintains a remarkable capacity to regenerate by compensatory growth. Specifically, in response to injury, quiescent hepatocytes enter the cell cycle and undergo DNA replication to promote liver regrowth. Despite the elucidation of a number of regenerative factors, the mechanisms by which liver injury triggers hepatocyte proliferation are incompletely understood. We demonstrate here that eosinophils stimulate liver regeneration after partial hepatectomy and toxin-mediated injury. Liver injury results in rapid recruitment of eosinophils, which secrete IL-4 to promote the proliferation of quiescent hepatocytes. Surprisingly, signaling via the IL-4Rα in macrophages, which have been implicated in tissue repair, is dispensable for hepatocyte proliferation and liver regrowth after injury. Instead, IL-4 exerts its proliferative actions via IL-4Rα in hepatocytes. Our findings thus provide a unique mechanism by which eosinophil-derived IL-4 stimulates hepatocyte proliferation in regenerating liver.


Asunto(s)
Eosinófilos/metabolismo , Interleucina-4/metabolismo , Regeneración Hepática/fisiología , Hígado/fisiología , Animales , Ciclo Celular/genética , Ciclo Celular/fisiología , Proliferación Celular , Perfilación de la Expresión Génica , Hepatectomía , Hepatocitos/citología , Hepatocitos/metabolismo , Hepatocitos/fisiología , Immunoblotting , Interleucina-4/genética , Subunidad alfa del Receptor de Interleucina-4/genética , Subunidad alfa del Receptor de Interleucina-4/metabolismo , Hígado/metabolismo , Hígado/cirugía , Regeneración Hepática/genética , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones Transgénicos , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Transducción de Señal/fisiología
5.
Nat Med ; 30(1): 265-270, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38177853

RESUMEN

The current third-line (and beyond) treatment options for RAS-mutant metastatic colorectal cancer have yielded limited efficacy. At the time of study start, the combination of sotorasib, a KRAS (Kirsten rat sarcoma viral oncogene homolog)-G12C inhibitor, and panitumumab, an epidermal growth factor receptor (EGFR) inhibitor, was hypothesized to overcome treatment-induced resistance. This phase 1b substudy of the CodeBreaK 101 master protocol evaluated sotorasib plus panitumumab in patients with chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. Here, we report the results in a dose-exploration cohort and a dose-expansion cohort. Patients received sotorasib (960 mg, once daily) plus panitumumab (6 mg kg-1, once every 2 weeks). The primary endpoints were safety and tolerability. Secondary endpoints included efficacy and pharmacokinetics. Exploratory biomarkers at baseline were assessed. Forty-eight patients (dose-exploration cohort, n = 8; dose-expansion cohort, n = 40) were treated. Treatment-related adverse events of any grade and grade ≥3 occurred in 45 (94%) and 13 (27%) patients, respectively. In the dose-expansion cohort, the confirmed objective response rate was 30.0% (95% confidence interval (CI) 16.6%, 46.5%). Median progression-free survival was 5.7 months (95% CI 4.2, 7.7 months). Median overall survival was 15.2 months (95% CI 12.5 months, not estimable). Prevalent genomic coalterations included APC (84%), TP53 (74%), SMAD4 (33%), PIK3CA (28%) and EGFR (26%). Sotorasib-panitumumab demonstrated acceptable safety with promising efficacy in chemotherapy-refractory KRASG12C-mutated metastatic colorectal cancer. ClinicalTrials.gov identifier: NCT04185883 .


Asunto(s)
Neoplasias Colorrectales , Piperazinas , Proteínas Proto-Oncogénicas p21(ras) , Piridinas , Pirimidinas , Humanos , Panitumumab/uso terapéutico , Proteínas Proto-Oncogénicas p21(ras)/genética , Proteínas Proto-Oncogénicas p21(ras)/metabolismo , Anticuerpos Monoclonales/efectos adversos , Neoplasias Colorrectales/tratamiento farmacológico , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/patología , Receptores ErbB , Mutación/genética , Protocolos de Quimioterapia Combinada Antineoplásica/efectos adversos
6.
Leuk Lymphoma ; : 1-11, 2024 May 07.
Artículo en Inglés | MEDLINE | ID: mdl-38712673

RESUMEN

AMG 330, a bispecific T-cell engager (BiTE®) that binds CD33 and CD3 on T cells facilitates T-cell-mediated cytotoxicity against CD33+ cells. This first-in-human, open-label, dose-escalation study evaluated the safety, pharmacokinetics, pharmacodynamics, and preliminary efficacy of AMG 330 in adults with relapsed/refractory acute myeloid leukemia (R/R AML). Amongst 77 patients treated with AMG 330 (0.5 µg/day-1.6 mg/day) on 14-day or 28-day cycles, maximum tolerated dose was not reached; median duration of treatment was 29 days. The most frequent treatment-related adverse events were cytokine release syndrome (CRS; 78%) and rash (30%); 10% of patients experienced grade 3/4 CRS. CRS was mitigated with stepwise dosing of AMG 330, prophylactic dexamethasone, and early treatment with tocilizumab. Among 60 evaluable patients, eight achieved complete remission or morphologic leukemia-free state; of the 52 non-responders, 37% had ≥50% reduction in AML bone marrow blasts. AMG 330 is a promising CD33-targeted therapeutic strategy for R/R AML.

7.
Nature ; 447(7148): 1116-20, 2007 Jun 28.
Artículo en Inglés | MEDLINE | ID: mdl-17515919

RESUMEN

Obesity and insulin resistance, the cardinal features of metabolic syndrome, are closely associated with a state of low-grade inflammation. In adipose tissue chronic overnutrition leads to macrophage infiltration, resulting in local inflammation that potentiates insulin resistance. For instance, transgenic expression of Mcp1 (also known as chemokine ligand 2, Ccl2) in adipose tissue increases macrophage infiltration, inflammation and insulin resistance. Conversely, disruption of Mcp1 or its receptor Ccr2 impairs migration of macrophages into adipose tissue, thereby lowering adipose tissue inflammation and improving insulin sensitivity. These findings together suggest a correlation between macrophage content in adipose tissue and insulin resistance. However, resident macrophages in tissues display tremendous heterogeneity in their activities and functions, primarily reflecting their local metabolic and immune microenvironment. While Mcp1 directs recruitment of pro-inflammatory classically activated macrophages to sites of tissue damage, resident macrophages, such as those present in the adipose tissue of lean mice, display the alternatively activated phenotype. Despite their higher capacity to repair tissue, the precise role of alternatively activated macrophages in obesity-induced insulin resistance remains unknown. Using mice with macrophage-specific deletion of the peroxisome proliferator activated receptor-gamma (PPARgamma), we show here that PPARgamma is required for maturation of alternatively activated macrophages. Disruption of PPARgamma in myeloid cells impairs alternative macrophage activation, and predisposes these animals to development of diet-induced obesity, insulin resistance, and glucose intolerance. Furthermore, gene expression profiling revealed that downregulation of oxidative phosphorylation gene expression in skeletal muscle and liver leads to decreased insulin sensitivity in these tissues. Together, our findings suggest that resident alternatively activated macrophages have a beneficial role in regulating nutrient homeostasis and suggest that macrophage polarization towards the alternative state might be a useful strategy for treating type 2 diabetes.


Asunto(s)
Resistencia a la Insulina/fisiología , Activación de Macrófagos , Macrófagos/citología , Macrófagos/metabolismo , PPAR gamma/metabolismo , Adiponectina/sangre , Tejido Adiposo/anatomía & histología , Tejido Adiposo/fisiología , Animales , Línea Celular , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/farmacología , Predisposición Genética a la Enfermedad , Glucosa/metabolismo , Homeostasis/efectos de los fármacos , Insulina/administración & dosificación , Insulina/metabolismo , Insulina/farmacología , Leishmania major/inmunología , Leishmania major/fisiología , Leishmaniasis Cutánea/inmunología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Tamaño de los Órganos/efectos de los fármacos , PPAR gamma/deficiencia , PPAR gamma/genética , Aumento de Peso/efectos de los fármacos
8.
Proc Natl Acad Sci U S A ; 107(52): 22617-22, 2010 Dec 28.
Artículo en Inglés | MEDLINE | ID: mdl-21149710

RESUMEN

Immune cells take residence in metabolic tissues, providing a framework for direct regulation of nutrient metabolism. Despite conservation of this anatomic relationship through evolution, the signals and mechanisms by which the immune system regulates nutrient homeostasis and insulin action remain poorly understood. Here, we demonstrate that the IL-4/STAT6 immune axis, a key pathway in helminth immunity and allergies, controls peripheral nutrient metabolism and insulin sensitivity. Disruption of signal transducer and activator of transcription 6 (STAT6) decreases insulin action and enhances a peroxisome proliferator-activated receptor α (PPARα) driven program of oxidative metabolism. Conversely, activation of STAT6 by IL-4 improves insulin action by inhibiting the PPARα-regulated program of nutrient catabolism and attenuating adipose tissue inflammation. These findings have thus identified an unexpected molecular link between the immune system and macronutrient metabolism, suggesting perhaps the coevolution of these pathways occurred to ensure access to glucose during times of helminth infection.


Asunto(s)
Metabolismo Energético/efectos de los fármacos , Resistencia a la Insulina/fisiología , Interleucina-4/farmacología , Factor de Transcripción STAT6/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Línea Celular , Células Cultivadas , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Femenino , Hepatocitos/citología , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Immunoblotting , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/metabolismo , Obesidad/etiología , Obesidad/metabolismo , Obesidad/fisiopatología , PPAR alfa/metabolismo , Fosforilación/efectos de los fármacos , Factor de Transcripción STAT6/genética , Transducción de Señal/efectos de los fármacos
9.
Cancer Res ; 83(8): 1175-1182, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-36625843

RESUMEN

Big data in healthcare can enable unprecedented understanding of diseases and their treatment, particularly in oncology. These data may include electronic health records, medical imaging, genomic sequencing, payor records, and data from pharmaceutical research, wearables, and medical devices. The ability to combine datasets and use data across many analyses is critical to the successful use of big data and is a concern for those who generate and use the data. Interoperability and data quality continue to be major challenges when working with different healthcare datasets. Mapping terminology across datasets, missing and incorrect data, and varying data structures make combining data an onerous and largely manual undertaking. Data privacy is another concern addressed by the Health Insurance Portability and Accountability Act, the Common Rule, and the General Data Protection Regulation. The use of big data is now included in the planning and activities of the FDA and the European Medicines Agency. The willingness of organizations to share data in a precompetitive fashion, agreements on data quality standards, and institution of universal and practical tenets on data privacy will be crucial to fully realizing the potential for big data in medicine.


Asunto(s)
Macrodatos , Neoplasias , Humanos , Neoplasias/diagnóstico , Neoplasias/terapia , Medicina de Precisión , Almacenamiento y Recuperación de la Información
10.
Cancer Res ; 83(8): 1183-1190, 2023 04 14.
Artículo en Inglés | MEDLINE | ID: mdl-36625851

RESUMEN

The analysis of big healthcare data has enormous potential as a tool for advancing oncology drug development and patient treatment, particularly in the context of precision medicine. However, there are challenges in organizing, sharing, integrating, and making these data readily accessible to the research community. This review presents five case studies illustrating various successful approaches to addressing such challenges. These efforts are CancerLinQ, the American Association for Cancer Research Project GENIE, Project Data Sphere, the National Cancer Institute Genomic Data Commons, and the Veterans Health Administration Clinical Data Initiative. Critical factors in the development of these systems include attention to the use of robust pipelines for data aggregation, common data models, data deidentification to enable multiple uses, integration of data collection into physician workflows, terminology standardization and attention to interoperability, extensive quality assurance and quality control activity, incorporation of multiple data types, and understanding how data resources can be best applied. By describing some of the emerging resources, we hope to inspire consideration of the secondary use of such data at the earliest possible step to ensure the proper sharing of data in order to generate insights that advance the understanding and the treatment of cancer.


Asunto(s)
Macrodatos , Neoplasias , Humanos , Estados Unidos/epidemiología , Neoplasias/genética , Neoplasias/terapia , Oncología Médica , Atención a la Salud
11.
Cell Metab ; 4(1): 13-24, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16814729

RESUMEN

Complex interplay between T helper (Th) cells and macrophages contributes to the formation and progression of atherosclerotic plaques. While Th1 cytokines promote inflammatory activation of lesion macrophages, Th2 cytokines attenuate macrophage-mediated inflammation and enhance their repair functions. In spite of its biologic importance, the biochemical and molecular basis of how Th2 cytokines promote maturation of anti-inflammatory macrophages is not understood. We show here that in response to interleukin-4 (IL-4), signal transducer and activator of transcription 6 (STAT6) and PPARgamma-coactivator-1beta (PGC-1beta) induce macrophage programs for fatty acid oxidation and mitochondrial biogenesis. Transgenic expression of PGC-1beta primes macrophages for alternative activation and strongly inhibits proinflammatory cytokine production, whereas inhibition of oxidative metabolism or RNAi-mediated knockdown of PGC-1beta attenuates this immune response. These data elucidate a molecular pathway that directly links mitochondrial oxidative metabolism to the anti-inflammatory program of macrophage activation, suggesting a potential role for metabolic therapies in treating atherogenic inflammation.


Asunto(s)
Metabolismo Energético/fisiología , Inflamación/metabolismo , Macrófagos/metabolismo , Transactivadores/metabolismo , Animales , Células Cultivadas , Ácidos Grasos/metabolismo , Retroalimentación Fisiológica/fisiología , Glucosa/metabolismo , Interferón gamma/farmacología , Interleucina-4/farmacología , Lipopolisacáridos/farmacología , Activación de Macrófagos , Macrófagos/efectos de los fármacos , Ratones , Ratones Transgénicos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Modelos Biológicos , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Factor de Transcripción STAT6/metabolismo , Transactivadores/efectos de los fármacos , Factores de Transcripción
12.
Clin Cancer Res ; 27(19): 5195-5212, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34321279

RESUMEN

The development of novel agents has transformed the treatment paradigm for multiple myeloma, with minimal residual disease (MRD) negativity now achievable across the entire disease spectrum. Bone marrow-based technologies to assess MRD, including approaches using next-generation flow and next-generation sequencing, have provided real-time clinical tools for the sensitive detection and monitoring of MRD in patients with multiple myeloma. Complementary liquid biopsy-based assays are now quickly progressing with some, such as mass spectrometry methods, being very close to clinical use, while others utilizing nucleic acid-based technologies are still developing and will prove important to further our understanding of the biology of MRD. On the regulatory front, multiple retrospective individual patient and clinical trial level meta-analyses have already shown and will continue to assess the potential of MRD as a surrogate for patient outcome. Given all this progress, it is not surprising that a number of clinicians are now considering using MRD to inform real-world clinical care of patients across the spectrum from smoldering myeloma to relapsed refractory multiple myeloma, with each disease setting presenting key challenges and questions that will need to be addressed through clinical trials. The pace of advances in targeted and immune therapies in multiple myeloma is unprecedented, and novel MRD-driven biomarker strategies are essential to accelerate innovative clinical trials leading to regulatory approval of novel treatments and continued improvement in patient outcomes.


Asunto(s)
Mieloma Múltiple , Médula Ósea , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mieloma Múltiple/diagnóstico , Mieloma Múltiple/tratamiento farmacológico , Neoplasia Residual/diagnóstico , Estudios Retrospectivos
13.
Cytometry B Clin Cytom ; 94(2): 239-249, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28475275

RESUMEN

BACKGROUND: Minimal residual disease (MRD) in B lymphoblastic leukemia (B-ALL) by flow cytometry is an established prognostic factor used to adjust treatment in most pediatric therapeutic protocols. MRD in B-ALL has been standardized by the Children's Oncology Group (COG) in North America, but not routine clinical labs. The Foundation for National Institutes of Health sought to harmonize MRD measurement among COG, oncology groups, academic, community and government, laboratories. METHODS: Listmode data from post-induction marrows were distributed from a reference lab to seven different clinical FCM labs with variable experience in B-ALL MRD. Labs were provided with the COG protocol. Files from 15 cases were distributed to the seven labs. Educational sessions were implemented, and 10 more listmode file cases analyzed. RESULTS: Among 105 initial challenges, the overall discordance rate was 26%. In the final round, performance improved considerably; out of 70 challenges, there were five false positives and one false negative (9% discordance), and no quantitative discordance. Four of six deviations occurred in a single lab. Three samples with hematogones were still misclassified as MRD. CONCLUSIONS: Despite the provision of the COG standardized analysis protocol, even experienced laboratories require an educational component for B-ALL MRD analysis by FCM. Recognition of hematogones remains challenging for some labs when using the COG protocol. The results from this study suggest that dissemination of MRD testing to other North American laboratories as part of routine clinical management of B-ALL is possible but requires additional educational components to complement standardized methodology. © 2017 International Clinical Cytometry Society.


Asunto(s)
Neoplasia Residual/diagnóstico , Neoplasia Residual/patología , Citometría de Flujo/métodos , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/diagnóstico , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Pronóstico
14.
JAMA Oncol ; 3(7): e170580, 2017 Jul 13.
Artículo en Inglés | MEDLINE | ID: mdl-28494052

RESUMEN

IMPORTANCE: Minimal residual disease (MRD) refers to the presence of disease in cases deemed to be in complete remission by conventional pathologic analysis. Assessing the association of MRD status following induction therapy in patients with acute lymphoblastic leukemia (ALL) with relapse and mortality may improve the efficiency of clinical trials and accelerate drug development. OBJECTIVE: To quantify the relationships between event-free survival (EFS) and overall survival (OS) with MRD status in pediatric and adult ALL using publications of clinical trials and other databases. DATA SOURCES: Clinical studies in ALL identified via searches of PubMed, MEDLINE, and clinicaltrials.gov. STUDY SELECTION: Our search and study screening process adhered to the PRISMA Guidelines. Studies that addressed EFS or OS by MRD status in patients with ALL were included; reviews, abstracts, and studies with fewer than 30 patients or insufficient MRD description were excluded. DATA EXTRACTION AND SYNTHESIS: Study sample size, patient age, follow-up time, timing of MRD assessment (postinduction or consolidation), MRD detection method, phenotype/genotype (B cell, T cell, Philadelphia chromosome), and EFS and OS. Searches of PubMed and MEDLINE identified 566 articles. A parallel search on clinicaltrials.gov found 67 closed trials and 62 open trials as of 2014. Merging results of 2 independent searches and applying exclusions gave 39 publications in 3 arms of patient populations (adult, pediatric, and mixed). We performed separate meta-analyses for each of these 3 subpopulations. RESULTS: The 39 publications comprised 13 637 patients: 16 adult studies (2076 patients), 20 pediatric (11 249 patients), and 3 mixed (312 patients). The EFS hazard ratio (HR) for achieving MRD negativity is 0.23 (95% Bayesian credible interval [BCI] 0.18-0.28) for pediatric patients and 0.28 (95% BCI, 0.24-0.33) for adults. The respective HRs in OS are 0.28 (95% BCI, 0.19-0.41) and 0.28 (95% BCI, 0.20-0.39). The effect was similar across all subgroups and covariates. CONCLUSIONS AND RELEVANCE: The value of having achieved MRD negativity is substantial in both pediatric and adult patients with ALL. These results are consistent across therapies, methods of and times of MRD assessment, cutoff levels, and disease subtypes. Minimal residual disease status warrants consideration as an early measure of disease response for evaluating new therapies, improving the efficiency of clinical trials, accelerating drug development, and for regulatory approval. A caveat is that an accelerated approval of a particular new drug using an intermediate end point, such as MRD, would require confirmation using traditional efficacy end points.


Asunto(s)
Neoplasia Residual/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Adulto , Niño , Humanos , Leucemia-Linfoma Linfoblástico de Células Precursoras/terapia , Pronóstico , Análisis de Supervivencia , Resultado del Tratamiento
15.
Clin Cancer Res ; 23(15): 3980-3993, 2017 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-28428191

RESUMEN

Treatment of myeloma has benefited from the introduction of more effective and better tolerated agents, improvements in supportive care, better understanding of disease biology, revision of diagnostic criteria, and new sensitive and specific tools for disease prognostication and management. Assessment of minimal residual disease (MRD) in response to therapy is one of these tools, as longer progression-free survival (PFS) is seen consistently among patients who have achieved MRD negativity. Current therapies lead to unprecedented frequency and depth of response, and next-generation flow and sequencing methods to measure MRD in bone marrow are in use and being developed with sensitivities in the range of 10-5 to 10-6 cells. These technologies may be combined with functional imaging to detect MRD outside of bone marrow. Moreover, immune profiling methods are being developed to better understand the immune environment in myeloma and response to immunomodulatory agents while methods for molecular profiling of myeloma cells and circulating DNA in blood are also emerging. With the continued development and standardization of these methodologies, MRD has high potential for use in gaining new drug approvals in myeloma. The FDA has outlined two pathways by which MRD could be qualified as a surrogate endpoint for clinical studies directed at obtaining accelerated approval for new myeloma drugs. Most importantly, better understanding of MRD should also contribute to better treatment monitoring. Potentially, MRD status could be used as a prognostic factor for making treatment decisions and for informing timing of therapeutic interventions. Clin Cancer Res; 23(15); 3980-93. ©2017 AACR.


Asunto(s)
ADN Tumoral Circulante/sangre , Mieloma Múltiple/sangre , Mieloma Múltiple/tratamiento farmacológico , Neoplasia Residual/sangre , Biomarcadores de Tumor/genética , Médula Ósea/efectos de los fármacos , Médula Ósea/patología , Supervivencia sin Enfermedad , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Mieloma Múltiple/complicaciones , Mieloma Múltiple/genética , Neoplasia Residual/inducido químicamente , Neoplasia Residual/genética , Selección de Paciente , Pronóstico
16.
Cell Signal ; 16(3): 375-84, 2004 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-14687667

RESUMEN

The interaction between CD40 ligand (CD154) expressed on activated T cells and its receptor, CD40, has been shown to play a role in the onset and maintenance of autoimmune inflammation. Recent studies suggest that CD154+T cells also contribute to the regulation of atherogenesis due to their capacity to activate CD40+cells of the vasculature, including vascular smooth muscle cells (VSMC). The present study evaluated the signalling events initiated through CD40 ligation which culminate in VSMC chemokine production. CD40 ligation resulted in the phosphorylation/activation of mitogen-activated protein kinases (MAPKs), extracellular signal-regulated kinases 1 and 2 (ERK1/2), and p38, but not c-jun N-terminal kinase. Inhibition of both ERK1/2 and p38 activity abrogated CD40 stimulation of IL-8 and MCP-1 production. CD40-mediated induction of chemokines also showed dependence on the Src family kinase activity. The Src kinase inhibitor, PP2, was found to inhibit CD40-induced phosphorylation of ERK1/2 as well as activation of IkappaB kinase. An evaluation of Src kinases that may be important in CD40 signalling identified Lyn as a potential candidate. These data indicate that CD40 signalling in VSMC activates a Src family kinase-initiated pathway that results in the induction of MAPK activities required for successful induction of chemokine synthesis.


Asunto(s)
Antígenos CD40/metabolismo , Quimiocina CCL2/biosíntesis , Quimiocina CCL5/biosíntesis , Sistema de Señalización de MAP Quinasas/fisiología , Músculo Liso Vascular/metabolismo , Familia-src Quinasas/metabolismo , Animales , Ligando de CD40/metabolismo , Células CHO , Técnicas de Cultivo de Célula , Cricetinae , Cricetulus , Activación Enzimática , Expresión Génica , Interleucina-8/biosíntesis , Ratones , Proteína Quinasa 3 Activada por Mitógenos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Monocitos/citología , Monocitos/metabolismo , Fosforilación , Transducción de Señal , Proteínas Quinasas p38 Activadas por Mitógenos
17.
Cell Host Microbe ; 14(2): 171-182, 2013 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-23954156

RESUMEN

Host-adapted Salmonella strains are responsible for a number of disease manifestations in mammals, including an asymptomatic chronic infection in which bacteria survive within macrophages located in systemic sites. However, the host cell physiology and metabolic requirements supporting bacterial persistence are poorly understood. In a mouse model of long-term infection, we found that S. typhimurium preferentially associates with anti-inflammatory/M2 macrophages at later stages of infection. Further, PPARδ, a eukaryotic transcription factor involved in sustaining fatty acid metabolism, is upregulated in Salmonella-infected macrophages. PPARδ deficiency dramatically inhibits Salmonella replication, which is linked to the metabolic state of macrophages and the level of intracellular glucose available to bacteria. Pharmacological activation of PPARδ increases glucose availability and enhances bacterial replication in macrophages and mice, while Salmonella fail to persist in Pparδ null mice. These data suggest that M2 macrophages represent a unique niche for long-term intracellular bacterial survival and link the PPARδ-regulated metabolic state of the host cell to persistent bacterial infection.


Asunto(s)
Interacciones Huésped-Patógeno , Macrófagos/microbiología , PPAR delta/metabolismo , Salmonella typhimurium/fisiología , Animales , Modelos Animales de Enfermedad , Glucosa/metabolismo , Ratones , Viabilidad Microbiana , Salmonelosis Animal , Salmonella typhimurium/crecimiento & desarrollo , Salmonella typhimurium/metabolismo
18.
J Exp Med ; 207(8): 1599-608, 2010 Aug 02.
Artículo en Inglés | MEDLINE | ID: mdl-20624891

RESUMEN

Peroxisome proliferator-activated receptors (PPARs; PPAR-alpha, PPAR-delta, and PPAR-gamma) comprise a family of nuclear receptors that sense fatty acid levels and translate this information into altered gene transcription. Previously, it was reported that treatment of mice with a synthetic ligand activator of PPAR-delta, GW0742, ameliorates experimental autoimmune encephalomyelitis (EAE), indicating a possible role for this nuclear receptor in the control of central nervous system (CNS) autoimmune inflammation. We show that mice deficient in PPAR-delta (PPAR-delta(-/-)) develop a severe inflammatory response during EAE characterized by a striking accumulation of IFN-gamma(+)IL-17A(-) and IFN-gamma(+)IL-17A(+) CD4(+) cells in the spinal cord. The preferential expansion of these T helper subsets in the CNS of PPAR-delta(-/-) mice occurred as a result of a constellation of immune system aberrations that included higher CD4(+) cell proliferation, cytokine production, and T-bet expression and enhanced expression of IL-12 family cytokines by myeloid cells. We also show that the effect of PPAR-delta in inhibiting the production of IFN-gamma and IL-12 family cytokines is ligand dependent and is observed in both mouse and human immune cells. Collectively, these findings suggest that PPAR-delta serves as an important molecular brake for the control of autoimmune inflammation.


Asunto(s)
Encefalomielitis Autoinmune Experimental/inmunología , Encefalomielitis Autoinmune Experimental/patología , PPAR delta/metabolismo , Linfocitos T Colaboradores-Inductores/patología , Animales , Encéfalo/inmunología , Encéfalo/patología , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/trasplante , Proliferación Celular , Encefalomielitis Autoinmune Experimental/metabolismo , Femenino , Expresión Génica/inmunología , Glicoproteínas/inmunología , Proteínas de Homeodominio/genética , Humanos , Interferón gamma/genética , Interferón gamma/metabolismo , Interleucina-12/genética , Interleucina-12/metabolismo , Interleucina-17/genética , Interleucina-17/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/inmunología , Leucocitos Mononucleares/metabolismo , Lipopolisacáridos/farmacología , Activación de Linfocitos/inmunología , Macrófagos Peritoneales/efectos de los fármacos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Glicoproteína Mielina-Oligodendrócito , Células Mieloides/efectos de los fármacos , Células Mieloides/inmunología , Células Mieloides/metabolismo , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , PPAR delta/antagonistas & inhibidores , Fragmentos de Péptidos/inmunología , Médula Espinal/inmunología , Médula Espinal/patología , Proteínas de Dominio T Box/genética , Linfocitos T Colaboradores-Inductores/efectos de los fármacos , Linfocitos T Colaboradores-Inductores/inmunología , Linfocitos T Colaboradores-Inductores/trasplante , Células TH1/inmunología , Células TH1/metabolismo , Tiazoles/farmacología , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
19.
Nat Med ; 15(11): 1266-72, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19838202

RESUMEN

Macrophages rapidly engulf apoptotic cells to limit the release of noxious cellular contents and to restrict autoimmune responses against self antigens. Although factors participating in recognition and engulfment of apoptotic cells have been identified, the transcriptional basis for the sensing and the silent disposal of apoptotic cells is unknown. Here we show that peroxisome proliferator-activated receptor-delta (PPAR-delta) is induced when macrophages engulf apoptotic cells and functions as a transcriptional sensor of dying cells. Genetic deletion of PPAR-delta decreases expression of opsonins such as complement component-1qb (C1qb), resulting in impairment of apoptotic cell clearance and reduction in anti-inflammatory cytokine production. This increases autoantibody production and predisposes global and macrophage-specific Ppard(-/-) mice to autoimmune kidney disease, a phenotype resembling the human disease systemic lupus erythematosus. Thus, PPAR-delta has a pivotal role in orchestrating the timely disposal of apoptotic cells by macrophages, ensuring that tolerance to self is maintained.


Asunto(s)
Apoptosis/fisiología , Autoinmunidad/fisiología , Tolerancia Inmunológica/inmunología , PPAR delta/metabolismo , Animales , Apoptosis/efectos de los fármacos , Autoanticuerpos/inmunología , Autoanticuerpos/metabolismo , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/patología , Enfermedades Autoinmunes/fisiopatología , Autoinmunidad/efectos de los fármacos , Antígeno CD11b/metabolismo , Células Cultivadas , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática/métodos , Femenino , Fluoresceínas , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/genética , Regulación de la Expresión Génica/inmunología , Humanos , Receptores de Hialuranos/metabolismo , Tolerancia Inmunológica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Proteínas Mitocondriales , Proteínas Opsoninas/genética , Proteínas Opsoninas/metabolismo , PPAR delta/agonistas , PPAR delta/deficiencia , PPAR delta/genética , Fagocitosis/efectos de los fármacos , Fagocitosis/inmunología , Regiones Promotoras Genéticas/efectos de los fármacos , Regiones Promotoras Genéticas/fisiología , ARN Mensajero/metabolismo , Tiazoles/farmacología , Timo/citología , Factores de Tiempo
20.
Cell Metab ; 7(6): 496-507, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18522831

RESUMEN

Macrophage infiltration and activation in metabolic tissues underlie obesity-induced insulin resistance and type 2 diabetes. While inflammatory activation of resident hepatic macrophages potentiates insulin resistance, the functions of alternatively activated Kupffer cells in metabolic disease remain unknown. Here we show that in response to the Th2 cytokine interleukin-4 (IL-4), peroxisome proliferator-activated receptor delta (PPARdelta) directs expression of the alternative phenotype in Kupffer cells and adipose tissue macrophages of lean mice. However, adoptive transfer of PPARdelta(-/-) (Ppard(-/-)) bone marrow into wild-type mice diminishes alternative activation of hepatic macrophages, causing hepatic dysfunction and systemic insulin resistance. Suppression of hepatic oxidative metabolism is recapitulated by treatment of primary hepatocytes with conditioned medium from PPARdelta(-/-) macrophages, indicating direct involvement of Kupffer cells in liver lipid metabolism. Taken together, these data suggest an unexpected beneficial role for alternatively activated Kupffer cells in metabolic syndrome and type 2 diabetes.


Asunto(s)
Resistencia a la Insulina , Macrófagos del Hígado/metabolismo , Activación de Macrófagos/fisiología , Macrófagos/metabolismo , PPAR delta/fisiología , Comunicación Paracrina , Tejido Adiposo/citología , Animales , Diabetes Mellitus Tipo 2 , Interleucina-4 , Macrófagos del Hígado/fisiología , Metabolismo de los Lípidos , Hígado/citología , Macrófagos/fisiología , Ratones , Obesidad/complicaciones
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA