Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
1.
Oncologist ; 27(7): 536-e553, 2022 07 05.
Artigo em Inglês | MEDLINE | ID: mdl-35325211

RESUMO

BACKGROUND: Patients with KRAS-mutant cancers have limited treatment options. Here we present a phase I study of JNJ-74699157, an oral, selective, covalent inhibitor of the KRAS G12C isoform, in patients with advanced cancer harboring the KRAS G12C mutation. METHODS: Eligible patients (aged ≥18 years) who had previously received or were ineligible for standard treatment received JNJ-74699157 once daily on a 21-day cycle. Dose escalation was guided by a modified continual reassessment method. RESULTS: Ten patients (100 mg: 9 and 200 mg: 1) were enrolled. Tumor types included non-small cell lung cancer (n = 5), colorectal cancer (n = 4), and carcinoma of unknown primary site (n = 1). The median age was 65 (range: 36-74) years and median treatment duration was 2.91 (range: 0.5-7.5) months. Dose-limiting toxicities of grades 3-4 increased blood creatinine phosphokinase (CPK) were observed in 100 mg and 200 mg dose levels. The most common adverse event was increased blood CPK (6 patients). No significant clinical benefit was observed; the best response was stable disease in 4 patients (40%). CONCLUSION: Based on dose-limiting skeletal muscle toxicities and the lack of efficacy at the 100 mg dose, further enrollment was stopped. The safety profile of JNJ-74699157 was not considered favorable for further clinical development. CLINICALTRIALS.GOV IDENTIFIER: NCT04006301.


Assuntos
Antineoplásicos , Carcinoma Pulmonar de Células não Pequenas , Neoplasias Pulmonares , Adolescente , Adulto , Idoso , Antineoplásicos/uso terapêutico , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Carcinoma Pulmonar de Células não Pequenas/genética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Mutação , Proteínas Proto-Oncogênicas p21(ras)/genética
2.
Clin Cancer Res ; 29(18): 3592-3602, 2023 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-37491846

RESUMO

PURPOSE: In this first-in-human, Phase 1, open-label, multicenter study, we evaluated JNJ-64619178, a selective and potent PRMT5 inhibitor, in patients with advanced malignant solid tumors or non-Hodgkin lymphomas (NHL). The primary objective was to evaluate the safety and to identify a recommended Phase 2 dose (RP2D) of JNJ-64619178. PATIENTS AND METHODS: Adult patients with treatment-refractory advanced solid tumors or NHL and measurable disease received escalating doses of JNJ-64619178 following two schedules (Schedule A: 14 days on/7 days off; Schedule B: every day on a 21-day cycle). Safety, pharmacokinetics (PK), pharmacodynamics (PD), and clinical activity were evaluated. RESULTS: Ninety patients received JNJ-64619178. Thrombocytopenia was identified as the only dose-limiting toxicity. JNJ-64619178 showed dose-proportional PK and robust target engagement, as measured by plasma symmetric dimethylarginine, across all dose levels. The objective response rate was 5.6% (5 of 90). Patients with adenoid cystic carcinoma (ACC) had an ORR of 11.5% (3 of 26) and a median progression-free survival of 19.1 months. CONCLUSIONS: JNJ-64619178 demonstrated manageable dose-dependent toxicity and preliminary evidence of antitumor activity in ACC and other tumor types. Plasma exposure was dose dependent, and target inhibition was maintained with intermittent and continuous dosing. On the basis of safety, clinical activity, PK, and PD findings, two provisional RP2Ds were selected: 1.5 mg intermittently and 1.0 mg once daily. Aside from ACC, clinical benefit was limited, and biomarkers to enrich for responsiveness to PRMT5 inhibition will be needed for further development.


Assuntos
Carcinoma Adenoide Cístico , Linfoma não Hodgkin , Neoplasias , Adulto , Humanos , Proteína-Arginina N-Metiltransferases/genética , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Pirimidinas , Pirróis
3.
Leuk Res ; 134: 107390, 2023 11.
Artigo em Inglês | MEDLINE | ID: mdl-37776843

RESUMO

Splicing factor (SF) gene mutations are frequent in myelodysplastic syndromes (MDS), and agents that modulate RNA splicing are hypothesized to provide clinical benefit. JNJ-64619178, a protein arginine methyltransferase 5 (PRMT5) inhibitor, was evaluated in patients with lower-risk (LR) MDS in a multi-part, Phase 1, multicenter study. The objectives were to determine a tolerable dose and to characterize safety, pharmacokinetics, pharmacodynamics, and preliminary clinical activity. JNJ-64619178 was administered on a 14 days on/7 days off schedule or every day on a 21-day cycle to patients with International Prognostic Scoring System (IPSS) Low or Intermediate-1 risk MDS who were red blood cell transfusion-dependent. Twenty-four patients were enrolled; 15 (62.5 %) patients had low IPSS risk score, while 18 (75.0 %) had an SF3B1 mutation. Median duration of treatment was 3.45 months (range: 0.03-6.93). No dose limiting toxicities were observed. The 0.5 mg once daily dose was considered better tolerated and chosen for dose expansion. Twenty-three (95.8 %) patients experienced treatment-emergent adverse events (TEAE). The most common TEAEs were neutropenia (15 [62.5 %]) and thrombocytopenia (14 [58.3 %]). JNJ-64619178 pharmacokinetics was dose-dependent. Target engagement as measured by plasma symmetric di-methylarginine was observed across all dose levels; however, variant allele frequency of clonal mutations in bone marrow or blood did not show sustained reductions from baseline. No patient achieved objective response or hematologic improvement per International Working Group 2006 criteria, or transfusion independence. A tolerable dose of JNJ-64619178 was identified in patients with LR MDS. However, no evidence of clinical benefit was observed.


Assuntos
Anemia , Síndromes Mielodisplásicas , Humanos , Proteína-Arginina N-Metiltransferases/genética , Proteína-Arginina N-Metiltransferases/uso terapêutico , Síndromes Mielodisplásicas/tratamento farmacológico , Síndromes Mielodisplásicas/genética , Anemia/tratamento farmacológico , Medula Óssea , Resultado do Tratamento
4.
J Immunol ; 182(9): 5183-7, 2009 May 01.
Artigo em Inglês | MEDLINE | ID: mdl-19380761

RESUMO

SLP-76 (Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa) organizes signaling from immunoreceptors, including the platelet collagen receptor, the pre-TCR, and the TCR, and is required for T cell development. In this study we examine a mouse in which wild-type SLP-76 is replaced with a mutant constitutively targeted to the cell membrane. Membrane-targeted SLP-76 (MTS) supports ITAM signaling in platelets and from the pre-TCR. Signaling from the mature TCR, however, is defective in MTS thymocytes, resulting in failed T cell differentiation. Defective thymic selection by MTS is not rescued by a SLP-76 mutant whose localization is restricted to the cytosol. Thus, fixed localization of SLP-76 reveals differential requirements for the subcellular localization of signaling complexes downstream of the pre-TCR vs mature TCR.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Proteínas Adaptadoras de Transdução de Sinal/genética , Membrana Celular/metabolismo , Glicoproteínas de Membrana/fisiologia , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Precursores de Proteínas/fisiologia , Receptores de Antígenos de Linfócitos T alfa-beta/fisiologia , Transdução de Sinais/imunologia , Linfócitos T/imunologia , Linfócitos T/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Animais , Diferenciação Celular/genética , Diferenciação Celular/imunologia , Membrana Celular/genética , Membrana Celular/imunologia , Técnicas de Introdução de Genes , Marcação de Genes , Humanos , Células Jurkat , Glicoproteínas de Membrana/biossíntese , Glicoproteínas de Membrana/genética , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fosfoproteínas/fisiologia , Precursores de Proteínas/biossíntese , Precursores de Proteínas/genética , Receptores de Antígenos de Linfócitos T alfa-beta/biossíntese , Receptores de Antígenos de Linfócitos T alfa-beta/genética , Transdução de Sinais/genética , Linfócitos T/citologia
5.
J Allergy Clin Immunol ; 124(5): 1088-98, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19895996

RESUMO

BACKGROUND: Lymphocyte cytosolic protein 2, also known as Src homology 2 domain-containing leukocyte phosphoprotein of 76 kilodaltons (SLP-76), is an essential adaptor molecule in myeloid cells, where it regulates FcepsilonRI-induced mast cell (MC) and FcgammaR- and integrin-induced neutrophil (polymorphonuclear leukocyte [PMN]) functions. SLP-76 contains 3 N-terminal tyrosines at residues 112, 128, and 145 that together are critical for its function. OBJECTIVE: We sought to explore the relative importance of tyrosines 112, 128, and 145 of SLP-76 during MC and PMN activation. METHODS: We examined in vitro MC and PMN functions using cells isolated from knock-in mice harboring phenylalanine substitution mutations at tyrosines 112 and 128 (Y112/128F) or 145 (Y145F). We also examined the effects of these mutations on in vivo MC and PMN activation using models of anaphylaxis, dermal inflammation, and serum-induced arthritis. RESULTS: Mutations at Y112/Y128 and Y145 both interfered with SLP-76 activity; however, Y145F had a greater effect than Y112/128F on most in vitro FcR-induced functions. In vitro functional defects were recapitulated in vivo, where mice expressing Y145F exhibited greater attenuation of MC-dependent passive systemic anaphylaxis and PMN-mediated inflammatory responses. Notably, the Y145F mutation completely protected mice against development of joint-specific inflammation in the MC and PMN-dependent K/B x N model of arthritis. CONCLUSION: Our data indicate that Y145 is the most critical tyrosine supporting SLP-76 function in myeloid cells. Future efforts to dissect how Y145 mediates SLP-76-dependent signaling in MCs and PMNs will increase our understanding of these lineages and provide insights into the treatment of allergy and inflammation.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/genética , Anafilaxia/genética , Artrite/genética , Dermatite/imunologia , Fosfoproteínas/genética , Anafilaxia/imunologia , Animais , Artrite/imunologia , Artrite/patologia , Dermatite/genética , Integrinas/imunologia , Mastócitos/imunologia , Mastócitos/metabolismo , Camundongos , Camundongos Mutantes , Mutação/genética , Mutação/imunologia , Neutrófilos/imunologia , Neutrófilos/metabolismo , Receptores de IgE/imunologia , Transdução de Sinais/imunologia , Tirosina/genética
6.
Neuro Oncol ; 20(12): 1634-1642, 2018 11 12.
Artigo em Inglês | MEDLINE | ID: mdl-29788497

RESUMO

Background: GSK2256098 is a novel oral focal adhesion kinase (FAK) inhibitor. Preclinical studies demonstrate growth inhibition in glioblastoma cell lines. However, rodent studies indicate limited blood-brain barrier (BBB) penetration. In this expansion cohort within a phase I study, the safety, tolerability, pharmacokinetics (PK), and clinical activity of GSK2256098 were evaluated in patients with recurrent glioblastoma. Biodistribution and kinetics of [11C]GSK2256098 were assessed in a substudy using positron-emission tomography (PET). Methods: Patients were treated with GSK2256098 until disease progression or withdrawal due to adverse events (AEs). Serial PK samples were collected on day 1. On a single day between days 9 and 20, patients received a microdose of intravenous [11C]GSK2256098 and were scanned with PET over 90 minutes with parallel PK sample collection. Response was assessed by MRI every 6 weeks. Results: Thirteen patients were treated in 3 dose cohorts (1000 mg, 750 mg, 500 mg; all dosed twice daily). The maximum tolerated dose was 1000 mg twice daily. Dose-limiting toxicities were related to cerebral edema. Treatment-related AEs (>25%) were diarrhea, fatigue, and nausea. Eight patients participated in the PET substudy, with [11C]GSK2256098 VT (volume of distribution) estimates of 0.9 in tumor tissue, 0.5 in surrounding T2 enhancing areas, and 0.4 in normal brain. Best response of stable disease was observed in 3 patients, including 1 patient on treatment for 11.3 months. Conclusions: GSK2256098 was tolerable in patients with relapsed glioblastoma. GSK2256098 crossed the BBB at low levels into normal brain, but at markedly higher levels into tumor, consistent with tumor-associated BBB disruption. Additional clinical trials of GSK2256098 are ongoing.


Assuntos
Aminopiridinas/uso terapêutico , Radioisótopos de Carbono/farmacocinética , Proteína-Tirosina Quinases de Adesão Focal/antagonistas & inibidores , Glioblastoma/tratamento farmacológico , Ácidos Hidroxâmicos/uso terapêutico , Recidiva Local de Neoplasia/tratamento farmacológico , Adolescente , Adulto , Idoso , Aminopiridinas/farmacocinética , Estudos de Coortes , Feminino , Seguimentos , Glioblastoma/patologia , Humanos , Ácidos Hidroxâmicos/farmacocinética , Masculino , Dose Máxima Tolerável , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/patologia , Prognóstico , Distribuição Tecidual , Adulto Jovem
7.
Exp Hematol ; 37(5): 549-58, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19375646

RESUMO

OBJECTIVE: In mice, homeostatic erythropoiesis occurs primarily in the bone marrow. However, in response to acute anemia, bone morphogenetic proteins 4 (BMP-4)-dependent stress erythropoiesis occurs in the adult spleen. BMP-4 can also regulate stress erythropoiesis in the fetal liver. In humans, erythropoiesis occurs in the bone marrow. However, in certain pathological conditions, extramedullary erythropoiesis is observed, where it can occur in several organs, including the liver. Given these observations, we propose to investigate whether the BMP-4-dependent stress erythropoiesis pathway can regulate extramedullary erythropoiesis in the livers of splenectomized mice. MATERIALS AND METHODS: Using splenectomized wild-type and flexed-tail (f) mice, which have a defect in BMP-4 signaling, we compared their recovery from phenylhydrazine-induced hemolytic anemia and characterized the expansion of stress burst-forming unit-erythroid in the livers of these mice during the recovery period. RESULTS: Our analysis indicates that in the absence of a spleen, stress erythropoiesis occurs in the murine liver. During the recovery, stress burst-forming unit-erythroid are expanded in the livers of splenectomized mice in response to BMP-4 expressed in the liver. f/f mice, which exhibit a defect in splenic stress erythropoiesis do not compensate for this defect by upregulating liver stress erythropoiesis. Furthermore, splenectomized f/f mice exhibit a defect in liver stress erythropoiesis, which demonstrates a role for the BMP-4-dependent stress erythropoiesis pathway in extramedullary erythropoiesis in the adult liver. CONCLUSIONS: Our data indicate that the BMP-4-dependent stress erythropoiesis pathway regulates extramedullary stress erythropoiesis, which occurs primarily in the murine spleen or in the case of splenectomized mice, in the adult liver.


Assuntos
Anemia Hemolítica/metabolismo , Proteína Morfogenética Óssea 4 , Hematopoese Extramedular , Fígado/metabolismo , Estresse Fisiológico , Anemia Hemolítica/induzido quimicamente , Anemia Hemolítica/genética , Anemia Hemolítica/patologia , Animais , Fígado/patologia , Camundongos , Camundongos Transgênicos , Oxidantes/toxicidade , Fenil-Hidrazinas/toxicidade , Esplenectomia
8.
Mamm Genome ; 18(12): 852-60, 2007 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18060457

RESUMO

Recent work has identified a growing body of evidence that subtle changes in noncoding sequences can result in significant pathology. These mutations, which would have been called silent polymorphisms in the past, affect gene transcription and mRNA splicing and lead to drastic changes in gene expression. Previous work from our lab has characterized the murine flexed-tail (f) mutation, which encodes Smad5, a transcription factor that functions downstream of the receptors for bone morphogenetic proteins (BMPs). f/f mice are unable to rapidly respond to acute anemia. Our analysis of these mice led to the development of a new model for stress erythropoiesis, where BMP4 expression in the spleen leads to the Smad5-dependent expansion of a specialized population of stress erythroid progenitors during the recovery from acute anemia. f/f mutant mice exhibit a defect in Smad5 mRNA splicing in the spleen such that the majority of Smad5 transcripts are two misspliced mRNAs. One of these mRNAs encodes a truncated form of Smad5 that inhibits BMP4 signaling when overexpressed. Here we show that a mutation in a poly(T) element in intron 4 causes the splicing defect in f/f mutant mice. This subtle mutation (loss of 1 or 2 Ts in a 16-T element) results in defects in splicing throughout the Smad5 gene. Furthermore, we show that this mutation results in tissue-specific splicing defects, which may explain why f/f mice are viable when Smad5-/- mice are embryonic lethal.


Assuntos
Íntrons , Mutação , Splicing de RNA/genética , Proteína Smad5/genética , Animais , Sequência de Bases , Mapeamento Cromossômico , Clonagem Molecular , Primers do DNA , Reparo do DNA , Amplificação de Genes , Camundongos , Camundongos Mutantes , Dados de Sequência Molecular
9.
J Immunol ; 178(7): 4606-14, 2007 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-17372019

RESUMO

The Src homology 2 domain-containing leukocyte phosphoprotein of 76 kDa (SLP-76) is an adaptor molecule critical for immunoreceptor and integrin signaling in multiple hemopoietic lineages. We showed previously that SLP-76 is required for neutrophil function in vitro, including integrin-induced adhesion and production of reactive oxygen intermediates, and to a lesser extent, FcgammaR-induced calcium flux and reactive oxygen intermediate production. It has been difficult to determine whether SLP-76 regulates neutrophil responses in vivo, because Slp-76(-/-) mice exhibit marked defects in thymocyte and vascular development, as well as platelet and mast cell function. To circumvent these issues, we generated mice with targeted loss of SLP-76 expression within myeloid cells. Neutrophils obtained from these animals failed to respond to integrin activation in vitro, similar to Slp-76(-/-) cells. Despite these abnormalities, SLP-76-deficient neutrophils migrated normally in vivo in response to Staphylococcus aureus infection and efficiently cleared micro-organisms. Interestingly, SLP-76-deficient neutrophils did not induce a robust inflammatory response in the localized Shwartzman reaction. Collectively, these data suggest that disruption of integrin signaling via loss of SLP-76 expression differentially impairs neutrophil functions in vivo, with preservation of migration and killing of S. aureus but reduction in LPS-induced tissue damage and vascular injury.


Assuntos
Abscesso/imunologia , Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Neutrófilos/imunologia , Fosfoproteínas/fisiologia , Fenômeno de Shwartzman/imunologia , Infecções Estafilocócicas/imunologia , Staphylococcus aureus , Abscesso/microbiologia , Abscesso/patologia , Proteínas Adaptadoras de Transdução de Sinal/genética , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Animais , Movimento Celular/genética , Integrases/genética , Integrinas/genética , Integrinas/metabolismo , Camundongos , Camundongos Mutantes , Células Mieloides/imunologia , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Fenômeno de Shwartzman/microbiologia , Fenômeno de Shwartzman/patologia , Transdução de Sinais , Infecções Estafilocócicas/patologia
10.
Blood ; 105(7): 2741-8, 2005 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-15591122

RESUMO

Acute anemia initiates a systemic response that results in the rapid mobilization and differentiation of erythroid progenitors in the adult spleen. The flexed-tail (f) mutant mice exhibit normal steady-state erythropoiesis but are unable to rapidly respond to acute erythropoietic stress. Here, we show that f/f mutant mice have a mutation in Madh5. Our analysis shows that BMP4/Madh5-dependent signaling, regulated by hypoxia, initiates the differentiation and expansion of erythroid progenitors in the spleen. These findings suggest a new model where stress erythroid progenitors, resident in the spleen, are poised to respond to changes in the microenvironment induced by acute anemia.


Assuntos
Anemia/fisiopatologia , Proteínas Morfogenéticas Ósseas/metabolismo , Proteínas de Ligação a DNA/metabolismo , Células Eritroides/citologia , Eritropoese/fisiologia , Fosfoproteínas/metabolismo , Transativadores/metabolismo , Doença Aguda , Alelos , Anemia/patologia , Animais , Proteína Morfogenética Óssea 4 , Proteínas Morfogenéticas Ósseas/genética , Diferenciação Celular/fisiologia , Proteínas de Ligação a DNA/genética , Células Eritroides/metabolismo , Células-Tronco Hematopoéticas/citologia , Células-Tronco Hematopoéticas/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Fosfoproteínas/genética , Transdução de Sinais/fisiologia , Proteína Smad5 , Baço/citologia , Transativadores/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA