Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 36
Filtrar
Más filtros

Banco de datos
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Nature ; 562(7728): 526-531, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30333627

RESUMEN

The implementation of targeted therapies for acute myeloid leukaemia (AML) has been challenging because of the complex mutational patterns within and across patients as well as a dearth of pharmacologic agents for most mutational events. Here we report initial findings from the Beat AML programme on a cohort of 672 tumour specimens collected from 562 patients. We assessed these specimens using whole-exome sequencing, RNA sequencing and analyses of ex vivo drug sensitivity. Our data reveal mutational events that have not previously been detected in AML. We show that the response to drugs is associated with mutational status, including instances of drug sensitivity that are specific to combinatorial mutational events. Integration with RNA sequencing also revealed gene expression signatures, which predict a role for specific gene networks in the drug response. Collectively, we have generated a dataset-accessible through the Beat AML data viewer (Vizome)-that can be leveraged to address clinical, genomic, transcriptomic and functional analyses of the biology of AML.


Asunto(s)
Regulación Neoplásica de la Expresión Génica/genética , Genoma Humano/genética , Genómica , Leucemia Mieloide Aguda/genética , Subunidad alfa 2 del Factor de Unión al Sitio Principal/genética , ADN (Citosina-5-)-Metiltransferasas/genética , ADN Metiltransferasa 3A , Conjuntos de Datos como Asunto , Exoma/genética , Femenino , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/metabolismo , Masculino , Terapia Molecular Dirigida , Proteínas Nucleares/genética , Nucleofosmina , Proteínas Proto-Oncogénicas/genética , Proteínas Represoras/genética , Análisis de Secuencia de ARN , Factores de Empalme Serina-Arginina/genética
2.
Proc Natl Acad Sci U S A ; 117(25): 14331-14341, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32513686

RESUMEN

Acute myeloid leukemia (AML) is the most common acute leukemia in adults, with approximately four new cases per 100,000 persons per year. Standard treatment for AML consists of induction chemotherapy with remission achieved in 50 to 75% of cases. Unfortunately, most patients will relapse and die from their disease, as 5-y survival is roughly 29%. Therefore, other treatment options are urgently needed. In recent years, immune-based therapies have led to unprecedented rates of survival among patients with some advanced cancers. Suppression of T cell function in the tumor microenvironment is commonly observed and may play a role in AML. We found that there is a significant association between T cell infiltration in the bone marrow microenvironment of newly diagnosed patients with AML and increased overall survival. Functional studies aimed at establishing the degree of T cell suppression in patients with AML revealed impaired T cell function in many patients. In most cases, T cell proliferation could be restored by blocking the immune checkpoint molecules PD-1, CTLA-4, or TIM3. Our data demonstrate that AML establishes an immune suppressive environment in the bone marrow, in part through T cell checkpoint function.


Asunto(s)
Médula Ósea/metabolismo , Leucemia Mieloide Aguda/metabolismo , Linfocitos T/metabolismo , Microambiente Tumoral/fisiología , Médula Ósea/inmunología , Antígeno CTLA-4/metabolismo , Proliferación Celular , Citocinas/metabolismo , Receptor 2 Celular del Virus de la Hepatitis A/metabolismo , Humanos , Leucemia Mieloide Aguda/inmunología , Leucemia Mieloide Aguda/terapia , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T/inmunología
3.
Blood ; 125(3): 504-15, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25370416

RESUMEN

Without effective therapy, chronic-phase chronic myeloid leukemia (CP-CML) evolves into an acute leukemia (blast crisis [BC]) that displays either myeloid or B-lymphoid characteristics. This transition is often preceded by a clinically recognized, but biologically poorly characterized, accelerated phase (AP). Here, we report that IKAROS protein is absent or reduced in bone marrow blasts from most CML patients with advanced myeloid disease (AP or BC). This contrasts with primitive CP-CML cells and BCR-ABL1-negative acute myeloid leukemia blasts, which express readily detectable IKAROS. To investigate whether loss of IKAROS contributes to myeloid disease progression in CP-CML, we examined the effects of forced expression of a dominant-negative isoform of IKAROS (IK6) in CP-CML patients' CD34(+) cells. We confirmed that IK6 disrupts IKAROS activity in transduced CP-CML cells and showed that it confers on them features of AP-CML, including a prolonged increased output in vitro and in xenografted mice of primitive cells with an enhanced ability to differentiate into basophils. Expression of IK6 in CD34(+) CP-CML cells also led to activation of signal transducer and activator of transcription 5 and transcriptional repression of its negative regulators. These findings implicate loss of IKAROS as a frequent step and potential diagnostic harbinger of progressive myeloid disease in CML patients.


Asunto(s)
Basófilos/patología , Diferenciación Celular/efectos de los fármacos , Eosinófilos/patología , Factor de Transcripción Ikaros/antagonistas & inhibidores , Leucemia Mieloide de Fase Crónica/patología , Factor de Transcripción STAT5/metabolismo , Animales , Antígenos CD34/metabolismo , Apoptosis/efectos de los fármacos , Basófilos/efectos de los fármacos , Basófilos/metabolismo , Western Blotting , Proliferación Celular/efectos de los fármacos , Células Cultivadas , Progresión de la Enfermedad , Eosinófilos/efectos de los fármacos , Eosinófilos/metabolismo , Citometría de Flujo , Humanos , Factor de Transcripción Ikaros/genética , Factor de Transcripción Ikaros/metabolismo , Técnicas para Inmunoenzimas , Leucemia Mieloide de Fase Crónica/genética , Leucemia Mieloide de Fase Crónica/metabolismo , Ratones , Ratones Endogámicos NOD , Ratones SCID , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Transcripción STAT5/genética
4.
Haematologica ; 102(11): 1890-1900, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28838991

RESUMEN

Although small molecule inhibitors of B-cell receptor-associated kinases have revolutionized therapy in chronic lymphocytic leukemia (CLL), responses are incomplete. Pro-survival signaling emanating from the microenvironment may foster therapeutic resistance of the malignant B cells resident in the protective lymphoid niches. B-cell activating factor (BAFF) is critical to the survival of both healthy and neoplastic B cells. However, the pro-survival pathways triggered by BAFF have not been fully characterized. Here we show that BAFF elicited resistance to spontaneous and drug-induced apoptosis in stromal co-cultures, induced activation of both canonical and non-canonical NFκB signaling pathways, and triggered B-cell receptor signaling in CLL cells, independently of IGHV mutational status. SYK, a proximal kinase in the B-cell receptor signaling cascade, acted via STAT3 to bolster transcription of the anti-apoptotic protein Mcl-1, thereby contributing to apoptosis resistance in BAFF-stimulated cells. SYK inhibitor entospletinib downregulated Mcl-1, abrogating BAFF-mediated cell survival. BAFF-B-cell receptor crosstalk in neoplastic B cells was mediated by SYK interaction with TRAF2/TRAF3 complex. Thus, SYK inhibition is a promising therapeutic strategy uniquely poised to antagonize crosstalk between BAFF and B-cell receptor, thereby disrupting the pro-survival microenvironment signaling in chronic lymphocytic leukemia.


Asunto(s)
Factor Activador de Células B/metabolismo , Leucemia Linfocítica Crónica de Células B/metabolismo , Proteína 1 de la Secuencia de Leucemia de Células Mieloides/metabolismo , Receptores de Antígenos de Linfocitos B/metabolismo , Transducción de Señal , Quinasa Syk/antagonistas & inhibidores , Animales , Células CHO , Línea Celular Tumoral , Supervivencia Celular/genética , Análisis por Conglomerados , Cricetulus , Perfilación de la Expresión Génica , Humanos , Cadenas Pesadas de Inmunoglobulina/genética , Inmunohistoquímica , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Leucemia Linfocítica Crónica de Células B/genética , Leucemia Linfocítica Crónica de Células B/patología , Modelos Biológicos , FN-kappa B/metabolismo , Unión Proteica , Proteínas Proto-Oncogénicas c-bcl-2/metabolismo , Factor 2 Asociado a Receptor de TNF/metabolismo , Factor 3 Asociado a Receptor de TNF
5.
N Engl J Med ; 368(19): 1781-90, 2013 May 09.
Artículo en Inglés | MEDLINE | ID: mdl-23656643

RESUMEN

BACKGROUND: The molecular causes of many hematologic cancers remain unclear. Among these cancers are chronic neutrophilic leukemia (CNL) and atypical (BCR-ABL1-negative) chronic myeloid leukemia (CML), both of which are diagnosed on the basis of neoplastic expansion of granulocytic cells and exclusion of genetic drivers that are known to occur in other myeloproliferative neoplasms and myeloproliferative-myelodysplastic overlap neoplasms. METHODS: To identify potential genetic drivers in these disorders, we used an integrated approach of deep sequencing coupled with the screening of primary leukemia cells obtained from patients with CNL or atypical CML against panels of tyrosine kinase-specific small interfering RNAs or small-molecule kinase inhibitors. We validated candidate oncogenes using in vitro transformation assays, and drug sensitivities were validated with the use of assays of primary-cell colonies. RESULTS: We identified activating mutations in the gene encoding the receptor for colony-stimulating factor 3 (CSF3R) in 16 of 27 patients (59%) with CNL or atypical CML. These mutations segregate within two distinct regions of CSF3R and lead to preferential downstream kinase signaling through SRC family-TNK2 or JAK kinases and differential sensitivity to kinase inhibitors. A patient with CNL carrying a JAK-activating CSF3R mutation had marked clinical improvement after the administration of the JAK1/2 inhibitor ruxolitinib. CONCLUSIONS: Mutations in CSF3R are common in patients with CNL or atypical CML and represent a potentially useful criterion for diagnosing these neoplasms. (Funded by the Leukemia and Lymphoma Society and others.).


Asunto(s)
Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/genética , Leucemia Neutrofílica Crónica/genética , Mutación , Receptores del Factor Estimulante de Colonias/genética , Animales , Humanos , Quinasas Janus/antagonistas & inhibidores , Leucemia Linfoide/genética , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/diagnóstico , Leucemia Neutrofílica Crónica/diagnóstico , Ratones , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Tirosina Quinasas/antagonistas & inhibidores , ARN Interferente Pequeño , Transducción de Señal/fisiología
6.
Blood ; 124(22): 3260-73, 2014 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-25293778

RESUMEN

Recent studies have revealed that p27, a nuclear cyclin-dependent kinase (Cdk) inhibitor and tumor suppressor, can acquire oncogenic activities upon mislocalization to the cytoplasm. To understand how these antagonistic activities influence oncogenesis, we dissected the nuclear and cytoplasmic functions of p27 in chronic myeloid leukemia (CML), a well-characterized malignancy caused by the BCR-ABL1 tyrosine kinase. p27 is predominantly cytoplasmic in CML and nuclear in normal cells. BCR-ABL1 regulates nuclear and cytoplasmic p27 abundance by kinase-dependent and -independent mechanisms, respectively. p27 knockdown in CML cell lines with predominantly cytoplasmic p27 induces apoptosis, consistent with a leukemogenic role of cytoplasmic p27. Accordingly, a p27 mutant (p27(CK-)) devoid of Cdk inhibitory nuclear functions enhances leukemogenesis in a murine CML model compared with complete absence of p27. In contrast, p27 mutations that enhance its stability (p27(T187A)) or nuclear retention (p27(S10A)) attenuate leukemogenesis over wild-type p27, validating the tumor-suppressor function of nuclear p27 in CML. We conclude that BCR-ABL1 kinase-dependent and -independent mechanisms convert p27 from a nuclear tumor suppressor to a cytoplasmic oncogene. These findings suggest that cytoplasmic mislocalization of p27 despite BCR-ABL1 inhibition by tyrosine kinase inhibitors may contribute to drug resistance, and effective therapeutic strategies to stabilize nuclear p27 must also prevent cytoplasmic mislocalization.


Asunto(s)
Transformación Celular Neoplásica/genética , Inhibidor p27 de las Quinasas Dependientes de la Ciclina/metabolismo , Citoplasma/metabolismo , Proteínas de Fusión bcr-abl/fisiología , Leucemia Mielógena Crónica BCR-ABL Positiva/genética , Animales , Células Cultivadas , Genes Supresores de Tumor , Humanos , Leucemia Mielógena Crónica BCR-ABL Positiva/patología , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Proteínas Oncogénicas/metabolismo , Transporte de Proteínas/genética
8.
Blood ; 122(22): 3628-31, 2013 Nov 21.
Artículo en Inglés | MEDLINE | ID: mdl-24081659

RESUMEN

We have recently identified targetable mutations in CSF3R (GCSFR) in 60% of chronic neutrophilic leukemia (CNL) and atypical (BCR-ABL-negative) chronic myeloid leukemia (aCML) patients. Here we demonstrate that the most prevalent, activating mutation, CSF3R T618I, is sufficient to drive a lethal myeloproliferative disorder in a murine bone marrow transplantation model. Mice transplanted with CSF3R T618I-expressing hematopoietic cells developed a myeloproliferative disorder characterized by overproduction of granulocytes and granulocytic infiltration of the spleen and liver, which was uniformly fatal. Treatment with the JAK1/2 inhibitor ruxolitinib lowered the white blood count and reduced spleen weight. This demonstrates that activating mutations in CSF3R are sufficient to drive a myeloproliferative disorder resembling aCML and CNL that is sensitive to pharmacologic JAK inhibition. This murine model is an excellent tool for the further study of neutrophilic myeloproliferative neoplasms and implicates the clinical use of JAK inhibitors for this disease.


Asunto(s)
Quinasas Janus/antagonistas & inhibidores , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Mutación Puntual , Pirazoles/uso terapéutico , Receptores del Factor Estimulante de Colonias/genética , Animales , Trasplante de Médula Ósea , Modelos Animales de Enfermedad , Granulocitos/patología , Humanos , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/tratamiento farmacológico , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/genética , Leucemia Neutrofílica Crónica/tratamiento farmacológico , Leucemia Neutrofílica Crónica/genética , Ratones , Ratones Endogámicos BALB C , Ratones Transgénicos , Trastornos Mieloproliferativos/patología , Neutrófilos/patología , Nitrilos , Inhibidores de Proteínas Quinasas/uso terapéutico , Pirimidinas
9.
Bioinformatics ; 29(4): 509-10, 2013 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-23303510

RESUMEN

SUMMARY: Determining the functional relevance of identified sequence variants in cancer is a prerequisite to ultimately matching specific therapies with individual patients. This level of mechanistic understanding requires integration of genomic information with complementary functional analyses to identify oncogenic targets and relies on the development of computational frameworks to aid in the prioritization and visualization of these diverse data types. In response to this, we have developed HitWalker, which prioritizes patient variants relative to their weighted proximity to functional assay results in a protein-protein interaction network. It is highly extensible, allowing incorporation of diverse data types to refine prioritization. In addition to a ranked list of variants, we have also devised a simple shortest path-based approach of visualizing the results in an intuitive manner to provide biological interpretation. AVAILABILITY AND IMPLEMENTATION: The program, documentation and example data are available as an R package from www.biodevlab.org/HitWalker.html.


Asunto(s)
Variación Genética , Neoplasias/genética , Programas Informáticos , Algoritmos , Genoma Humano , Genómica/métodos , Humanos , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/metabolismo , Mapeo de Interacción de Proteínas
10.
Mol Cell Proteomics ; 11(6): M112.017764, 2012 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-22345495

RESUMEN

Thymic stromal lymphopoietin (TSLP) is a cytokine that plays diverse roles in the regulation of immune responses. TSLP requires a heterodimeric receptor complex consisting of IL-7 receptor α subunit and its unique TSLP receptor (gene symbol CRLF2) to transmit signals in cells. Abnormal TSLP signaling (e.g. overexpression of TSLP or its unique receptor TSLPR) contributes to the development of a number of diseases including asthma and leukemia. However, a detailed understanding of the signaling pathways activated by TSLP remains elusive. In this study, we performed a global quantitative phosphoproteomic analysis of the TSLP signaling network using stable isotope labeling by amino acids in cell culture. By employing titanium dioxide in addition to antiphosphotyrosine antibodies as enrichment methods, we identified 4164 phosphopeptides on 1670 phosphoproteins. Using stable isotope labeling by amino acids in cell culture-based quantitation, we determined that the phosphorylation status of 226 proteins was modulated by TSLP stimulation. Our analysis identified activation of several members of the Src and Tec families of kinases including Btk, Lyn, and Tec by TSLP for the first time. In addition, we report TSLP-induced phosphorylation of protein phosphatases such as Ptpn6 (SHP-1) and Ptpn11 (Shp2), which has also not been reported previously. Co-immunoprecipitation assays showed that Shp2 binds to the adapter protein Gab2 in a TSLP-dependent manner. This is the first demonstration of an inducible protein complex in TSLP signaling. A kinase inhibitor screen revealed that pharmacological inhibition of PI-3 kinase, Jak family kinases, Src family kinases or Btk suppressed TSLP-dependent cellular proliferation making them candidate therapeutic targets in diseases resulting from aberrant TSLP signaling. Our study is the first phosphoproteomic analysis of the TSLP signaling pathway that greatly expands our understanding of TSLP signaling and provides novel therapeutic targets for TSLP/TSLPR-associated diseases in humans.


Asunto(s)
Citocinas/fisiología , Fosfoproteínas/metabolismo , Proteoma/metabolismo , Transducción de Señal , Secuencias de Aminoácidos , Animales , Línea Celular , Proliferación Celular/efectos de los fármacos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Marcaje Isotópico , Ratones , Fosfoproteínas Fosfatasas/metabolismo , Fosforilación , Unión Proteica , Mapas de Interacción de Proteínas , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Quinasas/metabolismo , Procesamiento Proteico-Postraduccional , Factor de Transcripción STAT3/metabolismo , Factor de Transcripción STAT5/metabolismo , Linfopoyetina del Estroma Tímico
11.
J Pharmacol Exp Ther ; 344(2): 378-87, 2013 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-23220742

RESUMEN

B-cell receptor (BCR) associated kinases including spleen tyrosine kinase (SYK) contribute to the pathogenesis of B-cell malignancies. SYK is persistently phosphorylated in a subset of non-Hodgkin lymphoma (NHL) and chronic lymphocytic leukemia (CLL), and SYK inhibition results in abrogation of downstream kinase activity and apoptosis. P505-15 (also known as PRT062607) is a novel, highly selective, and orally bioavailable small molecule SYK inhibitor (SYK IC(50) = 1 nM) with anti-SYK activity that is at least 80-fold greater than its affinity for other kinases. We evaluated the preclinical characteristics of P505-15 in models of NHL and CLL. P505-15 successfully inhibited SYK-mediated B-cell receptor signaling and decreased cell viability in NHL and CLL. Oral dosing in mice prevented BCR-mediated splenomegaly and significantly inhibited NHL tumor growth in a xenograft model. In addition, combination treatment of primary CLL cells with P505-15 plus fludarabine produced synergistic enhancement of activity at nanomolar concentrations. Our findings support the ongoing development of P505-15 as a therapeutic agent for B-cell malignancies. A dose finding study in healthy volunteers has been completed.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos B/efectos de los fármacos , Ciclohexilaminas/farmacología , Péptidos y Proteínas de Señalización Intracelular/antagonistas & inhibidores , Leucemia Linfocítica Crónica de Células B/tratamiento farmacológico , Linfoma no Hodgkin/tratamiento farmacológico , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Pirimidinas/farmacología , Vidarabina/análogos & derivados , Animales , Antineoplásicos/administración & dosificación , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapéutico , Apoptosis/efectos de los fármacos , Linfocitos B/enzimología , Linfocitos B/patología , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Ciclohexilaminas/administración & dosificación , Ciclohexilaminas/farmacocinética , Ciclohexilaminas/uso terapéutico , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Citometría de Flujo , Humanos , Leucemia Linfocítica Crónica de Células B/enzimología , Leucemia Linfocítica Crónica de Células B/patología , Linfoma no Hodgkin/enzimología , Linfoma no Hodgkin/patología , Ratones , Ratones Endogámicos BALB C , Ratones SCID , Fosforilación , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Pirimidinas/uso terapéutico , Bazo/efectos de los fármacos , Bazo/enzimología , Quinasa Syk , Vidarabina/administración & dosificación , Vidarabina/farmacocinética , Vidarabina/farmacología , Vidarabina/uso terapéutico , Ensayos Antitumor por Modelo de Xenoinjerto
12.
Blood ; 117(2): 591-4, 2011 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-20959606

RESUMEN

Phosphatidylinositol-3-kinase p110δ serves as a central integration point for signaling from cell surface receptors known to promote malignant B-cell proliferation and survival. This provides a rationale for the development of small molecule inhibitors that selectively target p110δ as a treatment approach for patients with B-cell malignancies. We thus identified 5-fluoro-3-phenyl-2-[(S)-1-(9H-purin-6-ylamino)-propyl]-3H-quinazolin-4-one (CAL-101), a highly selective and potent p110δ small molecule inhibitor (half-maximal effective concentration [EC(50)] = 8nM). Using tumor cell lines and primary patient samples representing multiple B-cell malignancies, we have demonstrated that constitutive phosphatidylinositol-3-kinase pathway activation is p110δ-dependent. CAL-101 blocked constitutive phosphatidylinositol-3-kinase signaling, resulting in decreased phosphorylation of Akt and other downstream effectors, an increase in poly(ADP-ribose) polymerase and caspase cleavage and an induction of apoptosis. These effects have been observed across a broad range of immature and mature B-cell malignancies, thereby providing a rationale for the ongoing clinical evaluation of CAL-101.


Asunto(s)
Antineoplásicos/farmacología , Leucemia de Células B/metabolismo , Linfoma de Células B/metabolismo , Fosfatidilinositol 3-Quinasas/efectos de los fármacos , Purinas/farmacología , Quinazolinonas/farmacología , Transducción de Señal/efectos de los fármacos , Animales , Western Blotting , Línea Celular Tumoral , Separación Celular , Supervivencia Celular/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Humanos , Ratones , Ratones Endogámicos C57BL , Fosfatidilinositol 3-Quinasas/metabolismo , Inhibidores de las Quinasa Fosfoinosítidos-3
13.
Blood ; 118(24): 6392-8, 2011 Dec 08.
Artículo en Inglés | MEDLINE | ID: mdl-21860020

RESUMEN

Proinflammatory cytokines such as TNFα are elevated in patients with myeloproliferative neoplasms (MPN), but their contribution to disease pathogenesis is unknown. Here we reveal a central role for TNFα in promoting clonal dominance of JAK2(V617F) expressing cells in MPN. We show that JAK2(V617F) kinase regulates TNFα expression in cell lines and primary MPN cells and TNFα expression is correlated with JAK2(V617F) allele burden. In clonogenic assays, normal controls show reduced colony formation in the presence of TNFα while colony formation by JAK2(V617F)-positive progenitor cells is resistant or stimulated by exposure to TNFα. Ectopic JAK2(V617F) expression confers TNFα resistance to normal murine progenitor cells and overcomes inherent TNFα hypersensitivity of Fanconi anemia complementation group C deficient progenitors. Lastly, absence of TNFα limits clonal expansion and attenuates disease in a murine model of JAK2(V617F)-positive MPN. Altogether our data are consistent with a model where JAK2(V617F) promotes clonal selection by conferring TNFα resistance to a preneoplastic TNFα sensitive cell, while simultaneously generating a TNFα-rich environment. Mutations that confer resistance to environmental stem cell stressors are a recognized mechanism of clonal selection and leukemogenesis in bone marrow failure syndromes and our data suggest that this mechanism is also critical to clonal selection in MPN.


Asunto(s)
Transformación Celular Neoplásica/metabolismo , Janus Quinasa 2/metabolismo , Trastornos Mieloproliferativos/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Sustitución de Aminoácidos , Animales , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Línea Celular Tumoral , Células Cultivadas , Proteína del Grupo de Complementación C de la Anemia de Fanconi/genética , Proteína del Grupo de Complementación C de la Anemia de Fanconi/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Humanos , Janus Quinasa 2/antagonistas & inhibidores , Janus Quinasa 2/genética , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/sangre , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/tratamiento farmacológico , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/genética , Leucemia Mieloide Crónica Atípica BCR-ABL Negativa/metabolismo , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , Ratones , Ratones Noqueados , Proteínas Mutantes/metabolismo , Células Progenitoras Mieloides/metabolismo , Trastornos Mieloproliferativos/sangre , Trastornos Mieloproliferativos/tratamiento farmacológico , Trastornos Mieloproliferativos/genética , Mutación Puntual , Inhibidores de Proteínas Quinasas/farmacología , ARN Mensajero/metabolismo , Proteínas Recombinantes/metabolismo , Factor de Necrosis Tumoral alfa/genética
14.
Pediatr Blood Cancer ; 60(8): 1313-9, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23526721

RESUMEN

BACKGROUND: Effective targeted therapies are needed in sarcomas, but the biological heterogeneity of these tumors has presented a challenge to clinical integration of small molecule inhibitors in sarcoma treatment. Here we outline a process to personalize therapy for sarcomas through a case study of a canine with spontaneous osteosarcoma. PROCEDURE: Rapid establishment of a primary tumor cell culture is described, followed by efficient functional characterization of the tumor that identified the Src inhibitor dasatinib as the most effective targeted therapy for this individual dog. RESULTS: Adjuvant dasatinib was administered for a total of 26 weeks following treatment with chemotherapy. Pharmacokinetic studies confirm that a therapeutic serum concentration was achieved at a tolerable dose of 0.75 mg/kg/day. The canine patient remains without evidence of recurrent disease 24 months following initial diagnosis. CONCLUSIONS: The approach described through this illustrative case study is broadly applicable and might be used for other solid tumors in canines as well as in humans.


Asunto(s)
Neoplasias Óseas , Enfermedades de los Perros/tratamiento farmacológico , Osteosarcoma , Inhibidores de Proteínas Quinasas , Pirimidinas , Tiazoles , Animales , Neoplasias Óseas/diagnóstico por imagen , Neoplasias Óseas/tratamiento farmacológico , Neoplasias Óseas/veterinaria , Línea Celular Tumoral , Dasatinib , Enfermedades de los Perros/diagnóstico por imagen , Perros , Osteosarcoma/diagnóstico por imagen , Osteosarcoma/tratamiento farmacológico , Osteosarcoma/veterinaria , Inhibidores de Proteínas Quinasas/administración & dosificación , Inhibidores de Proteínas Quinasas/farmacocinética , Pirimidinas/administración & dosificación , Pirimidinas/farmacocinética , Radiografía , Tiazoles/administración & dosificación , Tiazoles/farmacocinética , Factores de Tiempo , Familia-src Quinasas/antagonistas & inhibidores
15.
Biochem Biophys Res Commun ; 426(3): 363-8, 2012 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-22960170

RESUMEN

Alveolar rhabdomyosarcoma (aRMS) is a very aggressive sarcoma of children and young adults. Our previous studies have shown that small molecule inhibition of Pdgfra is initially very effective in an aRMS mouse model. However, slowly evolving, acquired resistance to a narrow-spectrum kinase inhibitor (imatinib) was common. We identified Src family kinases (SFKs) to be potentiators of Pdgfra in murine aRMS primary cell cultures from mouse tumors with evolved resistance in vivo in comparison to untreated cultures. Treating the resistant primary cell cultures with a combination of Pdgfra and Src inhibitors had a strong additive effect on cell viability. In Pdgfra knockout tumors, however, the Src inhibitor had no effect on tumor cell viability. Sorafenib, whose targets include not only PDGFRA but also the Src downstream target Raf, was effective at inhibiting mouse and human tumor cell growth and halted progression of mouse aRMS tumors in vivo. These results suggest that an adaptive Src-Pdgfra-Raf-Mapk axis is relevant to PDGFRA inhibition in rhabdomyosarcoma.


Asunto(s)
Resistencia a Antineoplásicos , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo , Rabdomiosarcoma Alveolar/enzimología , Rabdomiosarcoma Alveolar/patología , Quinasas raf/metabolismo , Familia-src Quinasas/metabolismo , Animales , Benzamidas , Bencenosulfonatos/farmacología , Proliferación Celular , Mesilato de Imatinib , Ratones , Niacinamida/análogos & derivados , Compuestos de Fenilurea , Piperazinas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/antagonistas & inhibidores , Sorafenib , Células Tumorales Cultivadas , Familia-src Quinasas/antagonistas & inhibidores
16.
Blood ; 115(25): 5232-40, 2010 Jun 24.
Artículo en Inglés | MEDLINE | ID: mdl-20385788

RESUMEN

Activating alleles of Janus kinase 2 (JAK2) such as JAK2(V617F) are central to the pathogenesis of myeloproliferative neoplasms (MPN), suggesting that small molecule inhibitors targeting JAK2 may be therapeutically useful. We have identified an aminopyrimidine derivative (CYT387), which inhibits JAK1, JAK2, and tyrosine kinase 2 (TYK2) at low nanomolar concentrations, with few additional targets. Between 0.5 and 1.5muM CYT387 caused growth suppression and apoptosis in JAK2-dependent hematopoietic cell lines, while nonhematopoietic cell lines were unaffected. In a murine MPN model, CYT387 normalized white cell counts, hematocrit, spleen size, and restored physiologic levels of inflammatory cytokines. Despite the hematologic responses and reduction of the JAK2(V617F) allele burden, JAK2(V617F) cells persisted and MPN recurred upon cessation of treatment, suggesting that JAK2 inhibitors may be unable to eliminate JAK2(V617F) cells, consistent with preliminary results from clinical trials of JAK2 inhibitors in myelofibrosis. While the clinical benefit of JAK2 inhibitors may be substantial, not the least due to reduction of inflammatory cytokines and symptomatic improvement, our data add to increasing evidence that kinase inhibitor monotherapy of malignant disease is not curative, suggesting a need for drug combinations to optimally target the malignant cells.


Asunto(s)
Apoptosis/efectos de los fármacos , Benzamidas/farmacología , Neoplasias Hematológicas/tratamiento farmacológico , Hematopoyesis/efectos de los fármacos , Janus Quinasa 2/antagonistas & inhibidores , Trastornos Mieloproliferativos/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/farmacología , Pirimidinas/farmacología , Animales , Apoptosis/genética , Apoptosis/inmunología , Línea Celular Tumoral , Citocinas/genética , Citocinas/inmunología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Ensayos de Selección de Medicamentos Antitumorales , Neoplasias Hematológicas/enzimología , Neoplasias Hematológicas/genética , Neoplasias Hematológicas/inmunología , Hematopoyesis/genética , Hematopoyesis/inmunología , Janus Quinasa 1/antagonistas & inhibidores , Janus Quinasa 1/genética , Janus Quinasa 1/inmunología , Janus Quinasa 1/metabolismo , Janus Quinasa 2/genética , Janus Quinasa 2/inmunología , Janus Quinasa 2/metabolismo , Ratones , Ratones Endogámicos BALB C , Mutación Missense , Trastornos Mieloproliferativos/enzimología , Trastornos Mieloproliferativos/genética , Trastornos Mieloproliferativos/inmunología
17.
Blood ; 115(2): 315-25, 2010 Jan 14.
Artículo en Inglés | MEDLINE | ID: mdl-19837975

RESUMEN

In chronic-phase chronic myeloid leukemia (CML) patients, the lack of a major cytogenetic response (< 36% Ph(+) metaphases) to imatinib within 12 months indicates failure and mandates a change of therapy. To identify biomarkers predictive of imatinib failure, we performed gene expression array profiling of CD34(+) cells from 2 independent cohorts of imatinib-naive chronic-phase CML patients. The learning set consisted of retrospectively selected patients with a complete cytogenetic response or more than 65% Ph(+) metaphases within 12 months of imatinib therapy. Based on analysis of variance P less than .1 and fold difference 1.5 or more, we identified 885 probe sets with differential expression between responders and nonresponders, from which we extracted a 75-probe set minimal signature (classifier) that separated the 2 groups. On application to a prospectively accrued validation set, the classifier correctly predicted 88% of responders and 83% of nonresponders. Bioinformatics analysis and comparison with published studies revealed overlap of classifier genes with CML progression signatures and implicated beta-catenin in their regulation, suggesting that chronic-phase CML patients destined to fail imatinib have more advanced disease than evident by morphologic criteria. Our classifier may allow directing more aggressive therapy upfront to the patients most likely to benefit while sparing good-risk patients from unnecessary toxicity.


Asunto(s)
Antígenos CD34/metabolismo , Antineoplásicos/administración & dosificación , Regulación Leucémica de la Expresión Génica/efectos de los fármacos , Leucemia Mielógena Crónica BCR-ABL Positiva/tratamiento farmacológico , Leucemia Mielógena Crónica BCR-ABL Positiva/metabolismo , Proteínas de Neoplasias/biosíntesis , Piperazinas/administración & dosificación , Pirimidinas/administración & dosificación , Adulto , Anciano , Benzamidas , Humanos , Mesilato de Imatinib , Masculino , Persona de Mediana Edad , Cromosoma Filadelfia/efectos de los fármacos
18.
J Vasc Interv Radiol ; 23(8): 1077-83, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22739646

RESUMEN

PURPOSE: To explore if addition of fibrinogen to the most commonly used experimental blood clot (EBC) model would improve its mechanical properties and histologic structure. MATERIALS AND METHODS: Fresh blood from three swine was used to create four EBC types. The Gralla model of thrombin-induced barium-opaque EBC served as the control. In three other EBC types, 50 mg, 100 mg, and 200 mg of bovine fibrinogen were added. Evaluation of EBCs was done with three tests: manual elongation, injection through an 8-F catheter, and an opacity test. Thirty EBCs of each type were evaluated with each test. Histologic evaluation followed. RESULTS: The control EBCs had low tensile strength and broke at 165% elongation. However, they were elastic and returned to their original length after catheter injection. The EBCs with fibrinogen exhibited increased tensile strength with increasing fibrinogen doses and withstood elongation to 213% (P < .01). Their elasticity decreased with increased tensile strength, and they remained elongated after catheter injection (P < .01 for EBC with 100 mg and 200 mg fibrinogen). Histologic examination showed more thorough mixing of blood with barium and a significantly increased amount of fibrin after addition of fibrinogen. CONCLUSIONS: Addition of fibrinogen to a Gralla EBC model changes its mechanical properties proportionately to the fibrinogen dose. Fibrinogen increases EBC tensile strength but decreases its elasticity. Fibrinogen also significantly increases the binding of blood cells with fibrin on histologic slides.


Asunto(s)
Coagulación Sanguínea , Fibrinógeno/metabolismo , Accidente Cerebrovascular/cirugía , Trombectomía/instrumentación , Animales , Adhesión Celular , Elasticidad , Diseño de Equipo , Eritrocitos/metabolismo , Ensayo de Materiales , Accidente Cerebrovascular/sangre , Porcinos , Resistencia a la Tracción , Trombina/metabolismo
19.
Proc Natl Acad Sci U S A ; 106(21): 8695-700, 2009 May 26.
Artículo en Inglés | MEDLINE | ID: mdl-19433805

RESUMEN

Targeted therapy has vastly improved outcomes in certain types of cancer. Extension of this paradigm across a broad spectrum of malignancies will require an efficient method to determine the molecular vulnerabilities of cancerous cells. Improvements in sequencing technology will soon enable high-throughput sequencing of entire genomes of cancer patients; however, determining the relevance of identified sequence variants will require complementary functional analyses. Here, we report an RNAi-assisted protein target identification (RAPID) technology that individually assesses targeting of each member of the tyrosine kinase gene family. We demonstrate that RAPID screening of primary leukemia cells from 30 patients identifies targets that are critical to survival of the malignant cells from 10 of these individuals. We identify known, activating mutations in JAK2 and K-RAS, as well as patient-specific sensitivity to down-regulation of FLT1, CSF1R, PDGFR, ROR1, EPHA4/5, JAK1/3, LMTK3, LYN, FYN, PTK2B, and N-RAS. We also describe a previously undescribed, somatic, activating mutation in the thrombopoietin receptor that is sensitive to down-stream pharmacologic inhibition. Hence, the RAPID technique can quickly identify molecular vulnerabilities in malignant cells. Combination of this technique with whole-genome sequencing will represent an ideal tool for oncogenic target identification such that specific therapies can be matched with individual patients.


Asunto(s)
Leucemia/genética , Leucemia/terapia , Interferencia de ARN , Alelos , Línea Celular Tumoral , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Janus Quinasa 2/genética , Janus Quinasa 2/metabolismo , Leucemia/metabolismo , Mutación/genética , Receptores de Trombopoyetina/genética , Factores de Tiempo , Resultado del Tratamiento
20.
Cancer Cell ; 40(8): 850-864.e9, 2022 08 08.
Artículo en Inglés | MEDLINE | ID: mdl-35868306

RESUMEN

Acute myeloid leukemia (AML) is a cancer of myeloid-lineage cells with limited therapeutic options. We previously combined ex vivo drug sensitivity with genomic, transcriptomic, and clinical annotations for a large cohort of AML patients, which facilitated discovery of functional genomic correlates. Here, we present a dataset that has been harmonized with our initial report to yield a cumulative cohort of 805 patients (942 specimens). We show strong cross-cohort concordance and identify features of drug response. Further, deconvoluting transcriptomic data shows that drug sensitivity is governed broadly by AML cell differentiation state, sometimes conditionally affecting other correlates of response. Finally, modeling of clinical outcome reveals a single gene, PEAR1, to be among the strongest predictors of patient survival, especially for young patients. Collectively, this report expands a large functional genomic resource, offers avenues for mechanistic exploration and drug development, and reveals tools for predicting outcome in AML.


Asunto(s)
Leucemia Mieloide Aguda , Diferenciación Celular , Estudios de Cohortes , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Receptores de Superficie Celular/genética , Transcriptoma
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA