Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 18 de 18
Filtrar
1.
Gynecol Oncol ; 172: 65-71, 2023 05.
Artículo en Inglés | MEDLINE | ID: mdl-36958197

RESUMEN

INTRODUCTION: Uterine leiomyosarcomas (uLMS) are rare, highly aggressive tumors. Up to 30% of uLMS may harbor gain of function (GOF) in the MAP2K4 gene, important for tumor cell proliferation, differentiation and metastasis. We investigated the in vivo activity of a novel MAP2K4 inhibitor, PLX8725, against uLMS harboring MAP2K4 gene-amplification. METHODS: Two fully characterized uLMS (i.e., LEY-11 and LEY-16) were grafted into female CB-17/SCID mice. Treatments with control vehicle or PLX8725 (50 mg/kg) were given via oral gavage daily on weekdays for up to 60 days. Tumor volume differences were calculated with two-way ANOVA. Pharmacokinetic (PK) and mechanistic studies of PLX8725 in uLMS PDX models were also performed. RESULTS: Both uLMS tumors evaluated demonstrated GOF in MAP2K4 (i.e., 3 CNV in both LEY-11 and LEY-16). Tumor growth inhibition was significantly greater in both PDX LEY-11 and PDX LEY-16 treated with PLX8725 when compared to controls (p < 0.001). Median overall survival was also significantly longer in both PDX LEY-11 (p = 0.0047) and PDX LEY-16 (p = 0.0058) treatment cohorts when compared to controls. PLX8725 oral treatment was well tolerated, and PK studies demonstrated that oral PLX8725 gives extended exposure in mice. Ex vivo tumor samples after PLX8725 exposure decreased phosphorylated-ATR, JNK and p38, and increased expression of apoptotic molecules on western blot. CONCLUSION: PLX8725 demonstrates promising in vivo activity against PDX models of uLMS harboring GOF alterations in the MAP2K4 gene with tolerable toxicity. Phase I trials of PLX8725 in advanced, recurrent, chemotherapy-resistant uLMS patients are warranted.


Asunto(s)
Leiomiosarcoma , Neoplasias Pélvicas , Neoplasias Uterinas , Humanos , Femenino , Animales , Ratones , Leiomiosarcoma/tratamiento farmacológico , Leiomiosarcoma/genética , Leiomiosarcoma/patología , Amplificación de Genes , Ratones SCID , Recurrencia Local de Neoplasia/genética , Neoplasias Uterinas/tratamiento farmacológico , Neoplasias Uterinas/genética , Neoplasias Uterinas/patología , MAP Quinasa Quinasa 4/genética
2.
Nature ; 526(7574): 583-6, 2015 Oct 22.
Artículo en Inglés | MEDLINE | ID: mdl-26466569

RESUMEN

Oncogenic activation of BRAF fuels cancer growth by constitutively promoting RAS-independent mitogen-activated protein kinase (MAPK) pathway signalling. Accordingly, RAF inhibitors have brought substantially improved personalized treatment of metastatic melanoma. However, these targeted agents have also revealed an unexpected consequence: stimulated growth of certain cancers. Structurally diverse ATP-competitive RAF inhibitors can either inhibit or paradoxically activate the MAPK pathway, depending whether activation is by BRAF mutation or by an upstream event, such as RAS mutation or receptor tyrosine kinase activation. Here we have identified next-generation RAF inhibitors (dubbed 'paradox breakers') that suppress mutant BRAF cells without activating the MAPK pathway in cells bearing upstream activation. In cells that express the same HRAS mutation prevalent in squamous tumours from patients treated with RAF inhibitors, the first-generation RAF inhibitor vemurafenib stimulated in vitro and in vivo growth and induced expression of MAPK pathway response genes; by contrast the paradox breakers PLX7904 and PLX8394 had no effect. Paradox breakers also overcame several known mechanisms of resistance to first-generation RAF inhibitors. Dissociating MAPK pathway inhibition from paradoxical activation might yield both improved safety and more durable efficacy than first-generation RAF inhibitors, a concept currently undergoing human clinical evaluation with PLX8394.


Asunto(s)
Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Animales , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Femenino , Genes ras/genética , Compuestos Heterocíclicos con 2 Anillos/efectos adversos , Compuestos Heterocíclicos con 2 Anillos/farmacología , Humanos , Indoles/efectos adversos , Indoles/farmacología , Sistema de Señalización de MAP Quinasas/genética , Ratones , Modelos Biológicos , Mutación/genética , Inhibidores de Proteínas Quinasas/efectos adversos , Proteínas Proto-Oncogénicas B-raf/genética , Sulfonamidas/efectos adversos , Sulfonamidas/farmacología , Vemurafenib
3.
N Engl J Med ; 373(5): 428-37, 2015 Jul 30.
Artículo en Inglés | MEDLINE | ID: mdl-26222558

RESUMEN

BACKGROUND: Expression of the colony-stimulating factor 1 (CSF1) gene is elevated in most tenosynovial giant-cell tumors. This observation has led to the discovery and clinical development of therapy targeting the CSF1 receptor (CSF1R). METHODS: Using x-ray co-crystallography to guide our drug-discovery research, we generated a potent, selective CSF1R inhibitor, PLX3397, that traps the kinase in the autoinhibited conformation. We then conducted a multicenter, phase 1 trial in two parts to analyze this compound. In the first part, we evaluated escalations in the dose of PLX3397 that was administered orally in patients with solid tumors (dose-escalation study). In the second part, we evaluated PLX3397 at the chosen phase 2 dose in an extension cohort of patients with tenosynovial giant-cell tumors (extension study). Pharmacokinetic and tumor responses in the enrolled patients were assessed, and CSF1 in situ hybridization was performed to confirm the mechanism of action of PLX3397 and that the pattern of CSF1 expression was consistent with the pathological features of tenosynovial giant-cell tumor. RESULTS: A total of 41 patients were enrolled in the dose-escalation study, and an additional 23 patients were enrolled in the extension study. The chosen phase 2 dose of PLX3397 was 1000 mg per day. In the extension study, 12 patients with tenosynovial giant-cell tumors had a partial response and 7 patients had stable disease. Responses usually occurred within the first 4 months of treatment, and the median duration of response exceeded 8 months. The most common adverse events included fatigue, change in hair color, nausea, dysgeusia, and periorbital edema; adverse events rarely led to discontinuation of treatment. CONCLUSIONS: Treatment of tenosynovial giant-cell tumors with PLX3397 resulted in a prolonged regression in tumor volume in most patients. (Funded by Plexxikon; ClinicalTrials.gov number, NCT01004861.).


Asunto(s)
Aminopiridinas/administración & dosificación , Tumores de Células Gigantes/tratamiento farmacológico , Pirroles/administración & dosificación , Receptor de Factor Estimulante de Colonias de Macrófagos/antagonistas & inhibidores , Neoplasias de los Tejidos Blandos/tratamiento farmacológico , Adulto , Anciano , Aminopiridinas/efectos adversos , Aminopiridinas/farmacocinética , Cristalografía por Rayos X , Relación Dosis-Respuesta a Droga , Descubrimiento de Drogas , Femenino , Tumores de Células Gigantes/patología , Humanos , Masculino , Persona de Mediana Edad , Pirroles/efectos adversos , Pirroles/farmacocinética , Receptor de Factor Estimulante de Colonias de Macrófagos/metabolismo , Neoplasias de los Tejidos Blandos/patología , Tendones/patología , Carga Tumoral
4.
Nature ; 467(7315): 596-9, 2010 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-20823850

RESUMEN

B-RAF is the most frequently mutated protein kinase in human cancers. The finding that oncogenic mutations in BRAF are common in melanoma, followed by the demonstration that these tumours are dependent on the RAF/MEK/ERK pathway, offered hope that inhibition of B-RAF kinase activity could benefit melanoma patients. Herein, we describe the structure-guided discovery of PLX4032 (RG7204), a potent inhibitor of oncogenic B-RAF kinase activity. Preclinical experiments demonstrated that PLX4032 selectively blocked the RAF/MEK/ERK pathway in BRAF mutant cells and caused regression of BRAF mutant xenografts. Toxicology studies confirmed a wide safety margin consistent with the high degree of selectivity, enabling Phase 1 clinical trials using a crystalline formulation of PLX4032 (ref. 5). In a subset of melanoma patients, pathway inhibition was monitored in paired biopsy specimens collected before treatment initiation and following two weeks of treatment. This analysis revealed substantial inhibition of ERK phosphorylation, yet clinical evaluation did not show tumour regressions. At higher drug exposures afforded by a new amorphous drug formulation, greater than 80% inhibition of ERK phosphorylation in the tumours of patients correlated with clinical response. Indeed, the Phase 1 clinical data revealed a remarkably high 81% response rate in metastatic melanoma patients treated at an oral dose of 960 mg twice daily. These data demonstrate that BRAF-mutant melanomas are highly dependent on B-RAF kinase activity.


Asunto(s)
Indoles/uso terapéutico , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Mutación/genética , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Sulfonamidas/uso terapéutico , Alelos , Animales , Perros , Quinasas MAP Reguladas por Señal Extracelular/antagonistas & inhibidores , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Indoles/administración & dosificación , Indoles/efectos adversos , Indoles/química , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Macaca fascicularis , Melanoma/genética , Melanoma/patología , Modelos Moleculares , Proteínas Mutantes/antagonistas & inhibidores , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Metástasis de la Neoplasia , Fosforilación/efectos de los fármacos , Tomografía de Emisión de Positrones , Proteínas Proto-Oncogénicas B-raf/química , Proteínas Proto-Oncogénicas B-raf/genética , Proteínas Proto-Oncogénicas B-raf/metabolismo , Ratas , Especificidad por Sustrato , Sulfonamidas/administración & dosificación , Sulfonamidas/efectos adversos , Sulfonamidas/química , Vemurafenib , Ensayos Antitumor por Modelo de Xenoinjerto
5.
Proc Natl Acad Sci U S A ; 110(14): 5689-94, 2013 Apr 02.
Artículo en Inglés | MEDLINE | ID: mdl-23493555

RESUMEN

Inflammation and cancer, two therapeutic areas historically addressed by separate drug discovery efforts, are now coupled in treatment approaches by a growing understanding of the dynamic molecular dialogues between immune and cancer cells. Agents that target specific compartments of the immune system, therefore, not only bring new disease modifying modalities to inflammatory diseases, but also offer a new avenue to cancer therapy by disrupting immune components of the microenvironment that foster tumor growth, progression, immune evasion, and treatment resistance. McDonough feline sarcoma viral (v-fms) oncogene homolog (FMS) and v-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog (KIT) are two hematopoietic cell surface receptors that regulate the development and function of macrophages and mast cells, respectively. We disclose a highly specific dual FMS and KIT kinase inhibitor developed from a multifaceted chemical scaffold. As expected, this inhibitor blocks the activation of macrophages, osteoclasts, and mast cells controlled by these two receptors. More importantly, the dual FMS and KIT inhibition profile has translated into a combination of benefits in preclinical disease models of inflammation and cancer.


Asunto(s)
Aminopiridinas/farmacología , Inflamación/tratamiento farmacológico , Modelos Moleculares , Metástasis de la Neoplasia/tratamiento farmacológico , Proteína Oncogénica gp140(v-fms)/antagonistas & inhibidores , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-kit/antagonistas & inhibidores , Pirroles/farmacología , Aminopiridinas/síntesis química , Aminopiridinas/química , Animales , Supervivencia Celular/efectos de los fármacos , Cromatografía de Afinidad , Cristalización , Escherichia coli , Células Endoteliales de la Vena Umbilical Humana , Humanos , Indoles , Macrófagos/efectos de los fármacos , Mastocitos/efectos de los fármacos , Estructura Molecular , Mutación Missense/genética , Proteína Oncogénica gp140(v-fms)/química , Proteína Oncogénica gp140(v-fms)/genética , Osteoclastos/efectos de los fármacos , Conformación Proteica , Inhibidores de Proteínas Quinasas/síntesis química , Inhibidores de Proteínas Quinasas/química , Proteínas Proto-Oncogénicas c-kit/química , Proteínas Proto-Oncogénicas c-kit/genética , Pirroles/síntesis química , Pirroles/química , Células Sf9 , Spodoptera
6.
N Engl J Med ; 366(3): 207-15, 2012 Jan 19.
Artículo en Inglés | MEDLINE | ID: mdl-22256804

RESUMEN

BACKGROUND: Cutaneous squamous-cell carcinomas and keratoacanthomas are common findings in patients treated with BRAF inhibitors. METHODS: We performed a molecular analysis to identify oncogenic mutations (HRAS, KRAS, NRAS, CDKN2A, and TP53) in the lesions from patients treated with the BRAF inhibitor vemurafenib. An analysis of an independent validation set and functional studies with BRAF inhibitors in the presence of the prevalent RAS mutation was also performed. RESULTS: Among 21 tumor samples, 13 had RAS mutations (12 in HRAS). In a validation set of 14 samples, 8 had RAS mutations (4 in HRAS). Thus, 60% (21 of 35) of the specimens harbored RAS mutations, the most prevalent being HRAS Q61L. Increased proliferation of HRAS Q61L-mutant cell lines exposed to vemurafenib was associated with mitogen-activated protein kinase (MAPK)-pathway signaling and activation of ERK-mediated transcription. In a mouse model of HRAS Q61L-mediated skin carcinogenesis, the vemurafenib analogue PLX4720 was not an initiator or a promoter of carcinogenesis but accelerated growth of the lesions harboring HRAS mutations, and this growth was blocked by concomitant treatment with a MEK inhibitor. CONCLUSIONS: Mutations in RAS, particularly HRAS, are frequent in cutaneous squamous-cell carcinomas and keratoacanthomas that develop in patients treated with vemurafenib. The molecular mechanism is consistent with the paradoxical activation of MAPK signaling and leads to accelerated growth of these lesions. (Funded by Hoffmann-La Roche and others; ClinicalTrials.gov numbers, NCT00405587, NCT00949702, NCT01001299, and NCT01006980.).


Asunto(s)
Carcinoma de Células Escamosas/genética , Genes ras , Indoles/uso terapéutico , Mutación , Inhibidores de Proteínas Quinasas/uso terapéutico , Proteínas Proto-Oncogénicas B-raf/antagonistas & inhibidores , Neoplasias Cutáneas/genética , Sulfonamidas/uso terapéutico , Anciano , Anciano de 80 o más Años , Animales , Carcinoma de Células Escamosas/tratamiento farmacológico , Femenino , Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Indoles/administración & dosificación , Masculino , Ratones , Persona de Mediana Edad , Quinasas de Proteína Quinasa Activadas por Mitógenos/metabolismo , Inhibidores de Proteínas Quinasas/administración & dosificación , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Sulfonamidas/administración & dosificación , Vemurafenib
7.
Phage (New Rochelle) ; 3(3): 171-178, 2022 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-36793550

RESUMEN

Background: Despite their importance to microbial dynamics involving Bacillus subtilis, we have a limited understanding of the diversity of phages that can lyse this model organism. Materials and Methods: Phages were isolated from soil samples collected from various sites in the southwest U.S. deserts on a wild B. subtilis strain. Their genomes were assembled, characterized, and bioinformatically compared. Results: Six Siphoviruses with high nucleotide and amino acid similarity to each other (>80%) but very limited similarity to phages currently in GenBank were isolated. These phages have double-stranded DNA genomes (55,312 to 56,127 bp) with 86-91 putative protein coding genes, and a low GC content. Comparative genomics reveal differences in loci encoding proteins that are putatively involved in bacterial adsorption with evidence for genomic mosaicism and a possible role for small genes. Conclusions: A comparative approach provides insights into phage evolution, including the role of indels in protein folding.

8.
JAMA Oncol ; 7(9): 1343-1350, 2021 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-34236401

RESUMEN

IMPORTANCE: Many cancer subtypes, including KIT-mutant gastrointestinal stromal tumors (GISTs), are driven by activating mutations in tyrosine kinases and may initially respond to kinase inhibitors but frequently relapse owing to outgrowth of heterogeneous subclones with resistance mutations. KIT inhibitors commonly used to treat GIST (eg, imatinib and sunitinib) are inactive-state (type II) inhibitors. OBJECTIVE: To assess whether combining a type II KIT inhibitor with a conformation-complementary, active-state (type I) KIT inhibitor is associated with broad mutation coverage and global disease control. DESIGN, SETTING, AND PARTICIPANTS: A highly selective type I inhibitor of KIT, PLX9486, was tested in a 2-part phase 1b/2a trial. Part 1 (dose escalation) evaluated PLX9486 monotherapy in patients with solid tumors. Part 2e (extension) evaluated PLX9486-sunitinib combination in patients with GIST. Patients were enrolled from March 2015 through February 2019; data analysis was performed from May 2020 through July 2020. INTERVENTIONS: Participants received 250, 350, 500, and 1000 mg of PLX9486 alone (part 1) or 500 and 1000 mg of PLX9486 together with 25 or 37.5 mg of sunitinib (part 2e) continuously in 28-day dosing cycles until disease progression, treatment discontinuation, or withdrawal. MAIN OUTCOMES AND MEASURES: Pharmacokinetics, safety, and tumor responses were assessed. Clinical efficacy end points (progression-free survival and clinical benefit rate) were supplemented with longitudinal monitoring of KIT mutations in circulating tumor DNA. RESULTS: A total of 39 PLX9486-naive patients (median age, 57 years [range, 39-79 years]; 22 men [56.4%]; 35 [89.7%] with refractory GIST) were enrolled in the dose escalation and extension parts. The recommended phase 2 dose of PLX9486 was 1000 mg daily. At this dose, PLX9486 could be safely combined with 25 or 37.5 mg daily of sunitinib continuously. Patients with GIST who received PLX9486 at a dose of 500 mg or less, at the recommended phase 2 dose, and with sunitinib had median (95% CI) progression-free survivals of 1.74 (1.54-1.84), 5.75 (0.99-11.0), and 12.1 (1.34-NA) months and clinical benefit rates (95% CI) of 14% (0%-58%), 50% (21%-79%), and 80% (52%-96%), respectively. CONCLUSIONS AND RELEVANCE: In this phase 1b/2a nonrandomized clinical trial, type I and type II KIT inhibitors PLX9486 and sunitinib were safely coadministered at the recommended dose of both single agents in patients with refractory GIST. Results suggest that cotargeting 2 complementary conformational states of the same kinase was associated with clinical benefit with an acceptable safety profile. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02401815.


Asunto(s)
Tumores del Estroma Gastrointestinal , Mesilato de Imatinib , Inhibidores de Proteínas Quinasas , Sunitinib , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/genética , Tumores del Estroma Gastrointestinal/patología , Humanos , Mesilato de Imatinib/efectos adversos , Masculino , Persona de Mediana Edad , Mutación , Recurrencia Local de Neoplasia/tratamiento farmacológico , Inhibidores de Proteínas Quinasas/efectos adversos , Sunitinib/efectos adversos
9.
Clin Exp Med ; 19(2): 201-210, 2019 May.
Artículo en Inglés | MEDLINE | ID: mdl-30523507

RESUMEN

The purpose of the present study was to investigate the in vitro and in vivo activity of PLX9486, a tyrosine kinase inhibitor (TKI) targeting both primary KIT exon 9 and 11 and secondary exon 17 and 18 mutations in gastrointestinal stromal tumors (GISTs). Imatinib, a potent inhibitor of mutated KIT, has revolutionized the clinical management of advanced, metastatic GIST. However, secondary resistance develops mainly through acquired mutations in KIT exons 13/14 or exons 17/18. Second-line sunitinib potently inhibits KIT exon 13/14 mutants but is ineffective against exon 17 mutations. In our study, PLX9486 demonstrated in vitro nanomolar potency in inhibiting the growth and KIT phosphorylation of engineered BaF3 cells transformed with KIT exon 17 mutations (p.D816V) and with the double KIT exon 11/17 mutations (p.V560G/D816V). The in vivo efficacy of PLX9486 was evaluated using two imatinib-resistant GIST patient-derived xenograft (PDX) models. In UZLX-GIST9 (KIT: p.P577del;W557LfsX5;D820G), PLX9486 100 mg/kg/day resulted in significant inhibition of proliferation. Pharmacodynamic analysis showed a pronounced reduction in mitogen-activated protein kinase (MAPK) activation and other downstream effects of the KIT signaling pathway but no significant effect on KIT Y703 and Y719 phosphorylation. Similarly, in MRL-GIST1 (KIT: p.W557_K558del;Y823D) PLX9486 treatment led to significant tumor regression and strong inhibition of MAPK activation. Interestingly, the inhibitory effect on MAPK activation was evident even after a single dose of PLX9486. In conclusion, PLX9486 showed anti-tumor efficacy in patient-derived imatinib-resistant GIST xenograft models, mainly through inhibition of KIT signaling. These preclinical efficacy data encourage further testing of PLX9486 in the clinical setting.


Asunto(s)
Antineoplásicos/administración & dosificación , Inhibidores Enzimáticos/administración & dosificación , Tumores del Estroma Gastrointestinal/tratamiento farmacológico , Tumores del Estroma Gastrointestinal/patología , Proteínas Mutantes/genética , Proteínas Tirosina Quinasas/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-kit/genética , Animales , Modelos Animales de Enfermedad , Evaluación Preclínica de Medicamentos , Xenoinjertos , Humanos , Ratones , Proteínas Mutantes/metabolismo , Trasplante de Neoplasias , Proteínas Proto-Oncogénicas c-kit/metabolismo , Resultado del Tratamiento
10.
Nat Commun ; 10(1): 3758, 2019 08 21.
Artículo en Inglés | MEDLINE | ID: mdl-31434879

RESUMEN

Many risk genes for the development of Alzheimer's disease (AD) are exclusively or highly expressed in myeloid cells. Microglia are dependent on colony-stimulating factor 1 receptor (CSF1R) signaling for their survival. We designed and synthesized a highly selective brain-penetrant CSF1R inhibitor (PLX5622) allowing for extended and specific microglial elimination, preceding and during pathology development. We find that in the 5xFAD mouse model of AD, plaques fail to form in the parenchymal space following microglial depletion, except in areas containing surviving microglia. Instead, Aß deposits in cortical blood vessels reminiscent of cerebral amyloid angiopathy. Altered gene expression in the 5xFAD hippocampus is also reversed by the absence of microglia. Transcriptional analyses of the residual plaque-forming microglia show they exhibit a disease-associated microglia profile. Collectively, we describe the structure, formulation, and efficacy of PLX5622, which allows for sustained microglial depletion and identify roles of microglia in initiating plaque pathogenesis.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Microglía/metabolismo , Compuestos Orgánicos/farmacología , Placa Amiloide/metabolismo , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/antagonistas & inhibidores , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/metabolismo , Enfermedad de Alzheimer/genética , Animales , Conducta Animal , Encéfalo/metabolismo , Modelos Animales de Enfermedad , Regulación de la Expresión Génica , Hipocampo/metabolismo , Humanos , Memoria , Ratones , Ratones Transgénicos , Receptores de Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Transcriptoma
11.
Mol Cell Biol ; 25(20): 8834-43, 2005 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-16199863

RESUMEN

Microbial pathogens have evolved diverse strategies to modulate the host cell cytoskeleton to achieve a productive infection and have proven instrumental for unraveling the molecular machinery that regulates actin polymerization. Here we uncover a mechanism for Shigella flexneri-induced actin comet tail elongation that links Abl family kinases to N-WASP-dependent actin polymerization. We show that the Abl kinases are required for Shigella actin comet tail formation, maximal intracellular motility, and cell-to-cell spread. Abl phosphorylates N-WASP, a host cell protein required for actin comet tail formation, and mutation of the Abl phosphorylation sites on N-WASP impairs comet tail elongation. Furthermore, we show that defective comet tail formation in cells lacking Abl kinases is rescued by activated forms of N-WASP. These data demonstrate for the first time that the Abl kinases play a role in the intracellular motility and intercellular dissemination of Shigella and uncover a new role for Abl kinases in the regulation of pathogen motility.


Asunto(s)
Actinas/química , Actinas/metabolismo , Proteínas Proto-Oncogénicas c-abl/metabolismo , Animales , Sitios de Unión , Células CACO-2 , Bovinos , Línea Celular , Células Cultivadas , Citoesqueleto/metabolismo , Humanos , Técnicas In Vitro , Ratones , Ratones Noqueados , Fosforilación , Proteínas Proto-Oncogénicas c-abl/deficiencia , Proteínas Proto-Oncogénicas c-abl/genética , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Shigella flexneri/patogenicidad , Tirosina/química
12.
Pain ; 156(9): 1692-1702, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25993548

RESUMEN

Tumor cells frequently metastasize to bone where they can generate cancer-induced bone pain (CIBP) that can be difficult to fully control using available therapies. Here, we explored whether PLX3397, a high-affinity small molecular antagonist that binds to and inhibits phosphorylation of colony-stimulating factor-1 receptor, the tyrosine-protein kinase c-Kit, and the FMS-like tyrosine kinase 3, can reduce CIBP. These 3 targets all regulate the proliferation and function of a subset of the myeloid cells including macrophages, osteoclasts, and mast cells. Preliminary experiments show that PLX3397 attenuated inflammatory pain after formalin injection into the hind paw of the rat. As there is an inflammatory component in CIBP, involving macrophages and osteoclasts, the effect of PLX3397 was explored in a prostate model of CIBP where skeletal pain, cancer cell proliferation, tumor metastasis, and bone remodeling could be monitored in the same animal. Administration of PLX3397 was initiated on day 14 after prostate cancer cell injection when the tumor was well established, and tumor-induced bone remodeling was first evident. Over the next 6 weeks, sustained administration of PLX3397 attenuated CIBP behaviors by approximately 50% and was equally efficacious in reducing tumor cell growth, formation of new tumor colonies in bone, and pathological tumor-induced bone remodeling. Developing a better understanding of potential effects that analgesic therapies have on the tumor itself may allow the development of therapies that not only better control the pain but also positively impact disease progression and overall survival in patients with bone cancer.


Asunto(s)
Aminopiridinas/uso terapéutico , Analgésicos/uso terapéutico , Neoplasias Óseas/complicaciones , Dolor/tratamiento farmacológico , Dolor/etiología , Pirroles/uso terapéutico , Animales , Antígenos CD/metabolismo , Antígenos de Diferenciación Mielomonocítica/metabolismo , Neoplasias Óseas/secundario , Huesos/patología , Línea Celular Tumoral , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Perros , Formaldehído/toxicidad , Masculino , Ratones , Ratones Desnudos , Trasplante de Neoplasias , Dimensión del Dolor/efectos de los fármacos , Neoplasias de la Próstata/patología , Proteínas Quinasas/metabolismo , Ratas , Ratas Sprague-Dawley
13.
Cancer Discov ; 5(6): 668-79, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25847190

RESUMEN

UNLABELLED: Tyrosine kinase domain mutations are a common cause of acquired clinical resistance to tyrosine kinase inhibitors (TKI) used to treat cancer, including the FLT3 inhibitor quizartinib. Mutation of kinase "gatekeeper" residues, which control access to an allosteric pocket adjacent to the ATP-binding site, has been frequently implicated in TKI resistance. The molecular underpinnings of gatekeeper mutation-mediated resistance are incompletely understood. We report the first cocrystal structure of FLT3 with the TKI quizartinib, which demonstrates that quizartinib binding relies on essential edge-to-face aromatic interactions with the gatekeeper F691 residue, and F830 within the highly conserved Asp-Phe-Gly motif in the activation loop. This reliance makes quizartinib critically vulnerable to gatekeeper and activation loop substitutions while minimizing the impact of mutations elsewhere. Moreover, we identify PLX3397, a novel FLT3 inhibitor that retains activity against the F691L mutant due to a binding mode that depends less vitally on specific interactions with the gatekeeper position. SIGNIFICANCE: We report the first cocrystal structure of FLT3 with a kinase inhibitor, elucidating the structural mechanism of resistance due to the gatekeeper F691L mutation. PLX3397 is a novel FLT3 inhibitor with in vitro activity against this mutation but is vulnerable to kinase domain mutations in the FLT3 activation loop.


Asunto(s)
Aminopiridinas/farmacología , Benzotiazoles/farmacología , Resistencia a Antineoplásicos/genética , Mutación , Compuestos de Fenilurea/farmacología , Pirroles/farmacología , Tirosina Quinasa 3 Similar a fms/genética , Aminopiridinas/química , Animales , Antineoplásicos/química , Antineoplásicos/farmacología , Benzotiazoles/química , Línea Celular Tumoral , Activación Enzimática/efectos de los fármacos , Xenoinjertos , Humanos , Leucemia Mieloide Aguda/tratamiento farmacológico , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/patología , Ratones , Modelos Moleculares , Conformación Molecular , Compuestos de Fenilurea/química , Unión Proteica , Dominios y Motivos de Interacción de Proteínas/genética , Inhibidores de Proteínas Quinasas/química , Inhibidores de Proteínas Quinasas/farmacología , Pirroles/química , Recurrencia , Relación Estructura-Actividad , Tirosina Quinasa 3 Similar a fms/química
14.
Surgery ; 135(6): 662-70, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15179373

RESUMEN

BACKGROUND: Three different protocols tested the hypothesis that hind limb muscle tissue O(2) saturation (StO(2)), measured noninvasively with near-infrared spectroscopy (NIRS), is as reliable as invasive systemic oxygenation indices to guide fluid resuscitation. METHODS: In series 1, swine (n=30) were hemorrhaged, then received either no fluid, a fixed volume of colloid (15 mL/kg), or shed blood plus lactated Ringer's (LR) titrated to MAP >60 mm Hg. In series 2, swine (n=16) received a penetrating femur injury, a 47% to 55% hemorrhage to determine a median lethal dose (LD(50)) then shed blood plus LR titrated to MAP >60 mm Hg. In series 3, swine (n=5) received the femur injury plus LD(50) hemorrhage, and were resuscitated with LR titrated to StO(2) >50%. RESULTS: In series 1, StO(2) tracked mixed venous O(2) saturation (SvO(2)), but discriminated between 3 survivor groups better than SvO(2), arterial lactate, or arterial base excess. In series 2, StO(2) tracked SvO(2) but discriminated between 2 survivor groups better than SvO(2), arterial lactate, or arterial base excess. In series 3, animals survived to extubation when resuscitated to an StO(2) target. CONCLUSIONS: Noninvasive muscle StO(2) determined by NIRS was more reliable than invasive oxygenation variables as an index of shock. Because muscle StO(2) can be easily monitored in emergency situations, it may represent an improved method to gauge the severity of shock or the adequacy of fluid resuscitation after trauma.


Asunto(s)
Fluidoterapia , Músculo Esquelético/metabolismo , Oxígeno/metabolismo , Resucitación , Choque Traumático/terapia , Animales , Arterias/metabolismo , Femenino , Fémur/lesiones , Fluidoterapia/normas , Miembro Posterior , Concentración de Iones de Hidrógeno , Ácido Láctico/sangre , Masculino , Resucitación/normas , Choque Traumático/diagnóstico , Espectroscopía Infrarroja Corta , Tasa de Supervivencia , Porcinos
15.
Pigment Cell Melanoma Res ; 26(6): 845-51, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-23890088

RESUMEN

BRAF is the most prevalent oncogene and an important therapeutic target in melanoma. In some cancers, BRAF is activated by rearrangements that fuse its kinase domain to 5' partner genes. We examined 848 comparative genomic hybridization profiles of melanocytic tumors and found copy number transitions within BRAF in 10 tumors, of which six could be further characterized by sequencing. In all, the BRAF kinase domain was fused in-frame to six N-terminal partners. No other mutations were identified in melanoma oncogenes. One of the seven melanoma cell lines without known oncogenic mutations harbored a similar BRAF fusion, which constitutively activated the MAP kinase pathway. Sorafenib, but not vemurafenib, could block MAP kinase pathway activation and proliferation of the cell line at clinically relevant concentrations, whereas BRAF(V) (600E) mutant melanoma cell lines were significantly more sensitive to vemurafenib. The patient from whom the cell line was derived showed a durable clinical response to sorafenib.


Asunto(s)
Melanocitos/patología , Melanoma/tratamiento farmacológico , Melanoma/enzimología , Terapia Molecular Dirigida , Proteínas de Fusión Oncogénica/metabolismo , Proteínas Proto-Oncogénicas B-raf/metabolismo , Neoplasias Cutáneas/enzimología , Adolescente , Adulto , Preescolar , Activación Enzimática/efectos de los fármacos , Femenino , Reordenamiento Génico/efectos de los fármacos , Humanos , Indoles/farmacología , Indoles/uso terapéutico , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Masculino , Melanocitos/efectos de los fármacos , Melanocitos/enzimología , Melanoma/patología , Persona de Mediana Edad , Nevo de Células Epitelioides y Fusiformes/patología , Niacinamida/análogos & derivados , Niacinamida/farmacología , Niacinamida/uso terapéutico , Compuestos de Fenilurea/farmacología , Compuestos de Fenilurea/uso terapéutico , Neoplasias Cutáneas/tratamiento farmacológico , Neoplasias Cutáneas/patología , Sorafenib , Sulfonamidas/farmacología , Sulfonamidas/uso terapéutico , Vemurafenib , Adulto Joven
16.
Appl Environ Microbiol ; 72(7): 5043-51, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16820504

RESUMEN

Shigellosis is a diarrheal disease caused by the gram-negative bacterium Shigella flexneri. Following ingestion of the bacterium, S. flexneri interferes with innate immunity, establishes an infection within the human colon, and initiates an inflammatory response that results in destruction of the tissue lining the gut. Examination of host cell factors required for S. flexneri pathogenesis in vivo has proven difficult due to limited host susceptibility. Here we report the development of a pathogenesis system that involves the use of Caenorhabditis elegans as a model organism to study S. flexneri virulence determinants and host molecules required for pathogenesis. We show that S. flexneri-mediated killing of C. elegans correlates with bacterial accumulation in the intestinal tract of the animal. The S. flexneri virulence plasmid, which encodes a type III secretory system as well as various virulence determinants crucial for pathogenesis in mammalian systems, was found to be required for maximal C. elegans killing. Additionally, we demonstrate that ABL-1, the C. elegans homolog of the mammalian c-Abl nonreceptor tyrosine kinase ABL1, is required for S. flexneri pathogenesis in nematodes. These data demonstrate the feasibility of using C. elegans to study S. flexneri pathogenesis in vivo and provide insight into host factors that contribute to S. flexneri pathogenesis.


Asunto(s)
Caenorhabditis elegans/enzimología , Caenorhabditis elegans/microbiología , Proteínas Proto-Oncogénicas c-abl/metabolismo , Shigella flexneri/patogenicidad , Animales , Intestinos/microbiología , Mutación , Plásmidos/genética , Proteínas Proto-Oncogénicas c-abl/genética , Shigella flexneri/genética , Virulencia/genética
17.
EMBO J ; 22(20): 5471-9, 2003 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-14532119

RESUMEN

Infection by the opportunistic bacterial pathogen Shigella flexneri stimulates tyrosine phosphorylation of host cell proteins, but the kinases involved and their effects on the regulation of cell signaling pathways during bacterial entry remain largely undefined. Here, we demonstrate a requirement for the Abl family of tyrosine kinases during Shigella internalization. Family members Abl and Arg are catalytically activated upon Shigella infection, accumulate at the site of bacterial entry, and are required for efficient bacterial uptake, as internalization is blocked upon targeted deletion of these kinases or treatment with a specific pharmacological inhibitor. We identify the adapter protein Crk as a target for Abl kinases during Shigella uptake, and show that a phosphorylation-deficient Crk mutant significantly inhibits bacterial uptake. Moreover, we define a novel signaling pathway activated during Shigella entry that links Abl kinase phosphorylation of Crk to activation of the Rho family GTPases Rac and Cdc42. Together, these findings reveal a new role for the Abl kinases, and suggest a novel approach to treatment of Shigella infections through inhibition of host cell signaling pathways.


Asunto(s)
Proteínas Proto-Oncogénicas c-abl/metabolismo , Shigella flexneri/enzimología , Shigella flexneri/patogenicidad , Animales , Arginina/deficiencia , Arginina/metabolismo , Células Cultivadas , Células HeLa , Humanos , Ratones , Ratones Noqueados , Proteínas Proto-Oncogénicas c-abl/deficiencia , Proteínas Proto-Oncogénicas c-abl/genética , Transducción de Señal
18.
J Trauma ; 57(3): 547-54, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15454801

RESUMEN

UNLABELLED: Hextend (HEX) is a colloid solution that is FDA-approved for volume expansion during surgery. ATL-146e is a novel adenosine A2A receptor agonist that has anti-inflammatory, neuroprotective, and coronary vasodilator properties. Three series of experiments were designed to evaluate the therapeutic potential of HEX+/-ATL-146e for emergency resuscitation from traumatic brain injury (TBI) + hemorrhagic hypotension. METHODS: In the first two studies in vivo, anesthetized, ventilated pigs (30-45 kg) received a fluid percussion TBI, 45% arterial hemorrhage, and 30 minutes shock period. In Series 1, resuscitation consisted of unlimited crystalloid (n = 8) or HEX (n = 8) to correct systolic arterial pressure >100 mm Hg and heart rate <100 bpm for the first 60 minutes ("emergency phase"), and then maintain cerebral perfusion pressure (CPP) > 70 mm Hg for 60-240 minutes. In Series 2 (n = 31), resuscitation consisted of a 1 L bolus of HEX + ATL-146e (10 ng/kg/min, n = 10) or HEX +placebo (n = 10) followed by crystalloid to the same endpoints. In Series 3 in vivo, the hemodynamic response evoked by 0, 10, 50, or 100 ng/kg/min ATL-146e was measured before or 60 minutes after HEX resuscitation from 45% hemorrhage. RESULTS: Following TBI+hemorrhage, there were 4/22 deaths in series 1 and 11/31 deaths in series 2. In those alive at 30 minutes, mean arterial pressure, cardiac index, mixed venous O2 saturation, and cerebral venous O2 saturation were all reduced by 40-60%, while heart rate and lactate were increased 2-5 fold. With no resuscitation (n = 2), there was minimal hemodynamic compensation and progressive acidosis. Upon resuscitation, these values corrected but intracranial pressure progressively rose from <5 mm Hg to 15-20 mm Hg. Series 1: With HEX (n = 8) versus crystalloid (n = 8), CPP was less labile, acid/base was maintained, and the fluid requirement was reduced by 60% (all p < 0.05) Series 2: With ATL-146e (n = 10) versus placebo (n = 10), stroke volume and cardiac output were improved by 40-60%, and the fluid requirement was reduced by 30% (all p < 0.05). Series 3: ATL-146e caused a dose-related increase (p < 0.05) in stroke volume after, but not before, hemorrhage. The effects on pre-load, afterload, and heart rate were similar before and after hemorrhage. CONCLUSIONS: HEX alone is a safe and efficacious low volume alternative to initial crystalloid resuscitation after TBI. An adenosine A2A agonist combined with 1 L of HEX safely and effectively counteracted a decrease in cardiac performance noted after TBI+hemorrhage without causing hypotension or bradycardia.


Asunto(s)
Lesiones Encefálicas/terapia , Ácidos Ciclohexanocarboxílicos/uso terapéutico , Derivados de Hidroxietil Almidón/uso terapéutico , Sustitutos del Plasma/uso terapéutico , Purinas/uso terapéutico , Resucitación/métodos , Animales , Presión Sanguínea/efectos de los fármacos , Gasto Cardíaco/efectos de los fármacos , Femenino , Masculino , Choque Hemorrágico/terapia , Volumen Sistólico/efectos de los fármacos , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA