Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 9 de 9
Filtrar
1.
Sci Signal ; 11(539)2018 07 17.
Artigo em Inglês | MEDLINE | ID: mdl-30018082

RESUMO

The protein tyrosine phosphatase PTPN11 is implicated in the pathogenesis of juvenile myelomonocytic leukemia (JMML), acute myeloid leukemia (AML), and other malignancies. Activating mutations in PTPN11 increase downstream proliferative signaling and cell survival. We investigated the signaling upstream of PTPN11 in JMML and AML cells and found that PTPN11 was activated by the nonreceptor tyrosine/serine/threonine kinase TNK2 and that PTPN11-mutant JMML and AML cells were sensitive to TNK2 inhibition. In cultured human cell-based assays, PTPN11 and TNK2 interacted directly, enabling TNK2 to phosphorylate PTPN11, which subsequently dephosphorylated TNK2 in a negative feedback loop. Mutations in PTPN11 did not affect this physical interaction but increased the basal activity of PTPN11 such that TNK2-mediated activation was additive. Consequently, coexpression of TNK2 and mutant PTPN11 synergistically increased mitogen-activated protein kinase (MAPK) signaling and enhanced colony formation in bone marrow cells from mice. Chemical inhibition of TNK2 blocked MAPK signaling and colony formation in vitro and decreased disease burden in a patient with PTPN11-mutant JMML who was treated with the multikinase (including TNK2) inhibitor dasatinib. Together, these data suggest that TNK2 is a promising therapeutic target for PTPN11-mutant leukemias.


Assuntos
Dasatinibe/farmacologia , Leucemia Mieloide Aguda/patologia , Leucemia Mielomonocítica Juvenil/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Proteínas Tirosina Quinases/antagonistas & inibidores , Mutações Sintéticas Letais , Animais , Criança , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Juvenil/tratamento farmacológico , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/genética , Masculino , Camundongos , Prognóstico , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais , Taxa de Sobrevida , Ensaio Tumoral de Célula-Tronco
2.
Dev Dyn ; 246(12): 1001-1014, 2017 12.
Artigo em Inglês | MEDLINE | ID: mdl-28975680

RESUMO

BACKGROUND: Accumulating evidence suggests the origin of juvenile myelomonocytic leukemia (JMML) is closely associated with fetal development. Nevertheless, the contribution of embryonic progenitors to JMML pathogenesis remains unexplored. We hypothesized that expression of JMML-initiating PTPN11 mutations in HSC-independent yolk sac erythromyeloid progenitors (YS EMPs) would result in a mouse model of pediatric myeloproliferative neoplasm (MPN). RESULTS: E9.5 YS EMPs from VavCre+;PTPN11D61Y embryos demonstrated growth hypersensitivity to granulocyte-macrophage colony-stimulating factor (GM-CSF) and hyperactive RAS-ERK signaling. Mutant EMPs engrafted the spleens of neonatal recipients, but did not cause disease. To assess MPN development during unperturbed hematopoiesis we generated CSF1R-MCM+;PTPN11E76K ;ROSAYFP mice in which oncogene expression was restricted to EMPs. Yellow fluorescent protein-positive progeny of mutant EMPs persisted in tissues one year after birth and demonstrated hyperactive RAS-ERK signaling. Nevertheless, these mice had normal survival and did not demonstrate features of MPN. CONCLUSIONS: YS EMPs expressing mutant PTPN11 demonstrate functional and molecular features of JMML but do not cause disease following transplantation nor following unperturbed development. Developmental Dynamics 246:1001-1014, 2017. © 2017 Wiley Periodicals, Inc.


Assuntos
Células Precursoras Eritroides/enzimologia , Mutação com Ganho de Função , Leucemia Mielomonocítica Juvenil/enzimologia , Sistema de Sinalização das MAP Quinases , Proteínas de Neoplasias/metabolismo , Células-Tronco Neoplásicas/enzimologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/metabolismo , Saco Vitelino/metabolismo , Animais , Células Precursoras Eritroides/patologia , Células Precursoras Eritroides/transplante , Leucemia Mielomonocítica Juvenil/embriologia , Leucemia Mielomonocítica Juvenil/genética , Leucemia Mielomonocítica Juvenil/patologia , Camundongos , Camundongos Transgênicos , Proteínas de Neoplasias/genética , Células-Tronco Neoplásicas/patologia , Proteína Tirosina Fosfatase não Receptora Tipo 11/genética , Saco Vitelino/patologia
3.
Pediatr Blood Cancer ; 62(4): 629-36, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25704135

RESUMO

BACKGROUND: Juvenile myelomonocytic leukemia (JMML) is not durably responsive to chemotherapy, and approximately 50% of patients relapse after hematopoietic stem cell transplant (HSCT). Here we report the activity and acute toxicity of the farnesyl transferase inhibitor tipifarnib, the response rate to 13-cis retinoic acid (CRA) in combination with cytoreductive chemotherapy, and survival following HSCT in children with JMML. PROCEDURE: Eighty-five patients with newly diagnosed JMML were enrolled on AAML0122 between 2001 and 2006. Forty-seven consented to receive tipifarnib in a phase II window before proceeding to a phase III trial of CRA in combination with fludarabine and cytarabine followed by HSCT and maintenance CRA. Thirty-eight patients enrolled only in the phase III trial. RESULTS: Overall response rate was 51% after tipifarnib and 68% after fludarabine/cytarabine/CRA. Tipifarnib did not increase pre-transplant toxicities. Forty-six percent of the 44 patients who received protocol compliant HSCT relapsed. Five-year overall survival was 55 ± 11% and event-free survival was 41 ± 11%, with no significant difference between patients who did or did not receive tipifarnib. CONCLUSIONS: Administration of tipifarnib in the window setting followed by HSCT in patients with newly diagnosed JMML was safe and yielded a 51% initial response rate as a single agent, but failed to reduce relapse rates or improve long-term overall survival.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Farnesil-Difosfato Farnesiltransferase/antagonistas & inibidores , Leucemia Mielomonocítica Juvenil/tratamento farmacológico , Quinolonas/administração & dosagem , Adulto , Idoso , Idoso de 80 Anos ou mais , Criança , Pré-Escolar , Citarabina/administração & dosagem , Intervalo Livre de Doença , Inibidores Enzimáticos/administração & dosagem , Feminino , Transplante de Células-Tronco Hematopoéticas , Humanos , Lactente , Isotretinoína/administração & dosagem , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/mortalidade , Leucemia Mielomonocítica Juvenil/patologia , Masculino , Pessoa de Meia-Idade , Taxa de Sobrevida , Vidarabina/administração & dosagem , Vidarabina/análogos & derivados
4.
J Clin Invest ; 124(6): 2762-73, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24812670

RESUMO

Overactive RAS signaling is prevalent in juvenile myelomonocytic leukemia (JMML) and the myeloproliferative variant of chronic myelomonocytic leukemia (MP-CMML) in humans, and both are refractory to conventional chemotherapy. Conditional activation of a constitutively active oncogenic Nras (NrasG12D/G12D) in murine hematopoietic cells promotes an acute myeloproliferative neoplasm (MPN) that recapitulates many features of JMML and MP-CMML. We found that NrasG12D/G12D-expressing HSCs, which serve as JMML/MP-CMML-initiating cells, show strong hyperactivation of ERK1/2, promoting hyperproliferation and depletion of HSCs and expansion of downstream progenitors. Inhibition of the MEK pathway alone prolonged the presence of NrasG12D/G12D-expressing HSCs but failed to restore their proper function. Consequently, approximately 60% of NrasG12D/G12D mice treated with MEK inhibitor alone died within 20 weeks, and the remaining animals continued to display JMML/MP-CMML-like phenotypes. In contrast, combined inhibition of MEK and JAK/STAT signaling, which is commonly hyperactivated in human and mouse CMML, potently inhibited human and mouse CMML cell growth in vitro, rescued mutant NrasG12D/G12D-expressing HSC function in vivo, and promoted long-term survival without evident disease manifestation in NrasG12D/G12D animals. These results provide a strong rationale for further exploration of combined targeting of MEK/ERK and JAK/STAT in treating patients with JMML and MP-CMML.


Assuntos
Janus Quinases/antagonistas & inibidores , Quinases de Proteína Quinase Ativadas por Mitógeno/antagonistas & inibidores , Transtornos Mieloproliferativos/tratamento farmacológico , Transtornos Mieloproliferativos/enzimologia , Animais , Proliferação de Células/efeitos dos fármacos , Genes ras , Humanos , Leucemia Mielomonocítica Crônica/tratamento farmacológico , Leucemia Mielomonocítica Crônica/enzimologia , Leucemia Mielomonocítica Crônica/genética , Leucemia Mielomonocítica Juvenil/tratamento farmacológico , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/genética , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Camundongos , Camundongos Mutantes , Transtornos Mieloproliferativos/patologia , Inibidores de Proteínas Quinases/administração & dosagem , Transdução de Sinais/efeitos dos fármacos
5.
J Clin Invest ; 124(4): 1794-809, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24569456

RESUMO

The genes encoding RAS family members are frequently mutated in juvenile myelomonocytic leukemia (JMML) and acute myeloid leukemia (AML). RAS proteins are difficult to target pharmacologically; therefore, targeting the downstream PI3K and RAF/MEK/ERK pathways represents a promising approach to treat RAS-addicted tumors. The p110α isoform of PI3K (encoded by Pik3ca) is an essential effector of oncogenic KRAS in murine lung tumors, but it is unknown whether p110α contributes to leukemia. To specifically examine the role of p110α in murine hematopoiesis and in leukemia, we conditionally deleted p110α in HSCs using the Cre-loxP system. Postnatal deletion of p110α resulted in mild anemia without affecting HSC self-renewal; however, deletion of p110α in mice with KRASG12D-associated JMML markedly delayed their death. Furthermore, the p110α-selective inhibitor BYL719 inhibited growth factor-independent KRASG12D BM colony formation and sensitized cells to a low dose of the MEK inhibitor MEK162. Furthermore, combined inhibition of p110α and MEK effectively reduced proliferation of RAS-mutated AML cell lines and disease in an AML murine xenograft model. Together, our data indicate that RAS-mutated myeloid leukemias are dependent on the PI3K isoform p110α, and combined pharmacologic inhibition of p110α and MEK could be an effective therapeutic strategy for JMML and AML.


Assuntos
Genes ras , Hematopoese/genética , Hematopoese/fisiologia , Leucemia Mieloide Aguda/enzimologia , Leucemia Mieloide Aguda/genética , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/genética , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Animais , Linhagem Celular Tumoral , Classe I de Fosfatidilinositol 3-Quinases , Eritropoese/genética , Eritropoese/fisiologia , Xenoenxertos , Humanos , Leucemia Mieloide Aguda/patologia , Leucemia Mielomonocítica Juvenil/patologia , Sistema de Sinalização das MAP Quinases , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fosfatidilinositol 3-Quinases/deficiência , Transdução de Sinais
6.
Artigo em Inglês | MEDLINE | ID: mdl-21239819

RESUMO

Expansion of myeloid blasts with suppression of normal hematopoiesis is a hallmark of acute myeloid leukemia (AML). In contrast, myeloproliferative neoplasms (MPNs) are clonal disorders characterized by overproliferation of one or more lineages that retain the ability to differentiate. Juvenile myelomonocytic leukemia (JMML) is an aggressive MPN of childhood that is clinically characterized by the overproduction of monocytic cells that can infiltrate organs, including the spleen, liver, gastrointestinal tract, and lung. Major progress in understanding the pathogenesis of JMML has been achieved by mapping out the genetic lesions that occur in patients. The spectrum of mutations described thus far in JMML occur in genes that encode proteins that signal through the Ras/mitogen-activated protein kinase (MAPK) pathways, thus providing potential new opportunities for both diagnosis and therapy. These genes include NF1, NRAS, KRAS, PTPN11, and, most recently, CBL. While the current standard of care for patients with JMML relies on allogeneic hematopoietic stem-cell transplant, relapse is the most frequent cause of treatment failure. Rarely, spontaneous resolution of this disorder can occur but is unpredictable. This review is focused on the genetic abnormalities that occur in JMML, with particular attention to germ-line predisposition syndromes associated with the disorder. Current approaches to therapy are also discussed.


Assuntos
Leucemia Mielomonocítica Juvenil/diagnóstico , Leucemia Mielomonocítica Juvenil/terapia , Síndromes Mielodisplásicas/diagnóstico , Síndromes Mielodisplásicas/terapia , Criança , Transplante de Células-Tronco Hematopoéticas , Humanos , Leucemia Mielomonocítica Juvenil/enzimologia , Leucemia Mielomonocítica Juvenil/genética , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Prognóstico
8.
Acta Haematol ; 119(4): 207-11, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18566538

RESUMO

Juvenile myelomonocytic leukemia (JMML) is a rare blood cell malignancy occurring in very young children. Yet, just as has been proven in other rare diseases, the study of JMML has provided us great insights into aberrant and dysregulated signal transduction through the Ras pathway, with the ultimate development of malignancy. Further, JMML investigations have also revealed to us much about the genetic predisposition to cancer.


Assuntos
Leucemia Mielomonocítica Juvenil/enzimologia , Transdução de Sinais , Proteínas ras/metabolismo , Criança , Pré-Escolar , Humanos , Leucemia Mielomonocítica Juvenil/genética , Transdução de Sinais/genética , Proteínas ras/genética
9.
Haematologica ; 92(11): 1557-60, 2007 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-18024405

RESUMO

FLT3 gene mutations have been identified as prognostic factors in myeloid malignancies. Furthermore, FLT3 can be activated by wild type overexpression or ligand-dependent in leukemic cells co-expressing FLT3 ligand (FLT3L). So far no data are available on FLT3/FLT3L expression and activation in JMML. In 51 clinical JMML samples, activating mutations were screened, FLT3 and FLT3L mRNA levels were assessed and the sensitivity of JMML cells to the FLT3 inhibitor PKC412 was tested by MTT assays. No evidence for constitutively activation of FLT3/FLT3L was found in JMML, indicating that FLT3 inhibitors are unlikely to be effective in JMML.


Assuntos
Leucemia Mielomonocítica Juvenil/etiologia , Tirosina Quinase 3 Semelhante a fms/fisiologia , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Humanos , Leucemia Mielomonocítica Juvenil/enzimologia , RNA Mensageiro/análise , Estaurosporina/análogos & derivados , Estaurosporina/farmacologia , Tirosina Quinase 3 Semelhante a fms/antagonistas & inibidores , Tirosina Quinase 3 Semelhante a fms/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...