Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Toxicol Pathol ; 44(1): 98-111, 2016 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-26534939

RESUMO

The growth plate, ovary, adrenal gland, and rodent incisor tooth are sentinel organs for antiangiogenic effects since they respond reliably, quantitatively, and sensitively to inhibition of the vascular endothelial growth factor receptor (VEGFR). Here we report that treatment of rats with platelet-derived growth factor receptor beta (PDGFRß) inhibitors that target pericytes results in severe ovarian hemorrhage with degeneration and eventual rupture of the corpus luteum. Evaluation of the growth plate, adrenal gland, and incisor tooth that are typical target organs for antiangiogenic treatment in the rodent revealed no abnormalities. Histologically, the changes in the ovary were characterized by sinusoidal dilatation, increased vessel fragility, and hemorrhage into the corpus luteum. Immunocytochemical staining of vessels with alpha smooth muscle actin and CD31 that recognize pericytes and vascular endothelium, respectively, demonstrated that this effect was due to selective pericyte deficiency within corpora lutea. Further experiments in which rats were treated concurrently with both PDGFRß and VEGFR inhibitors ablated the hemorrhagic response, resulting instead in corpus luteum necrosis. These changes are consistent with the notion that selective pericyte loss in the primitive capillary network resulted in increased vessel fragility and hemorrhage, whereas concomitant VEGFR inhibition resulted in vessel regression and reduced vascular perfusion that restricted development of the hemorrhagic vessels. These results also highlight the utility of the rodent ovary to respond differentially to VEGFR and PDGFR inhibitors, which may provide useful information during routine safety assessment for determining target organ toxicity.


Assuntos
Corpo Lúteo/efeitos dos fármacos , Hemorragia/induzido quimicamente , Ovário/efeitos dos fármacos , Pericitos/efeitos dos fármacos , Receptor beta de Fator de Crescimento Derivado de Plaquetas/antagonistas & inibidores , Actinas/metabolismo , Animais , Corpo Lúteo/fisiopatologia , Feminino , Histocitoquímica , Ovário/patologia , Ovário/fisiopatologia , Molécula-1 de Adesão Celular Endotelial a Plaquetas/metabolismo , Inibidores de Proteínas Quinases/farmacologia , Ratos , Ratos Wistar
2.
Nurse Res ; 22(1): 27-31, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25251817

RESUMO

AIM: To reflect on the author's personal and professional journey when undertaking semi-structured interviews on sensitive topics with potentially vulnerable people. BACKGROUND: When discussing care at the end of life, researchers must accept that some participants may become distressed or emotional, depending on their previous experiences. Interviews that involve sensitive topics require careful planning. DATA SOURCES: The semi-structured interviews were conducted as part of the author's PhD study examining the experiences of advance care planning among family caregivers of people with advanced dementia. REVIEW METHODS: A reflection on my personal and professional journey when undertaking semi-structured interviews on sensitive topics with potentially vulnerable people. DISCUSSION: The frustration and tragedy of dementia, as experienced by the family caregivers, were powerful and required the author to exert self-control to avoid being overly sympathetic and offering words of reassurance, agreement and comfort. CONCLUSION: This blurring of roles between researcher and nurse has implications for all nurse researchers who undertake qualitative interviews, particularly when an intense emotional response is likely. IMPLICATIONS FOR RESEARCH/PRACTICE: Nurse researchers should plan and prepare for potential blurring of roles during emotional interviews and should never automatically assume that they are sufficiently prepared as a result of their previous experience and nurse training.


Assuntos
Entrevistas como Assunto , Pesquisa em Enfermagem , Pesquisadores , Empatia , Humanos , Recursos Humanos
3.
Int J Cancer ; 129(4): 847-58, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21328342

RESUMO

Neutrophils are important innate immune cells that are involved in microbial clearance at sites of infection and in wound healing. The microenvironment of tumors often resembles that of chronic inflammation and increased numbers of neutrophils have been observed in several tumors and, in some cases, these positively correlate with poor prognosis. Neutrophil recruitment into tumors appears to be dependent on chemokines that bind to CXCR1 and CXCR2 expressed by neutrophils. In our study, we used lung adenocarcinoma A549 multicellular tumor spheroids and A549 tumor xenografts along with a CXCR2-specific small molecule inhibitor (AZ10397767) to investigate the recruitment and function of human neutrophils in tumors. We found that A549 spheroids constitutively secrete high levels of CXCL chemokines and that neutrophil recruitment into A549 tumors in vitro and in vivo is largely dependent on CXCR2 activation. AZ10397767 significantly reduced the numbers of infiltrating neutrophils into both in vitro and in vivo tumor models, which was associated with slower growing tumors. Neutrophil infiltration into A549 tumor spheroids increased their size compared to noninfiltrated spheroids and neutrophil-derived factors increased the proliferation of A549 tumor cells and induced endothelial cell tubule formation in vitro. In contrast, we saw no reduction in microvascular density in AZ10397767-treated A549 tumors or in tumors grown in CXCR2(-/-) mice, suggesting that angiogenesis in these tumors is CXCR2-independent. Our data show that neutrophils can contribute to lung tumor growth and that CXCR2 antagonists may be a useful therapeutic agent in the treatment of lung carcinomas.


Assuntos
Adenocarcinoma/tratamento farmacológico , Neoplasias Pulmonares/tratamento farmacológico , Melanoma/tratamento farmacológico , Infiltração de Neutrófilos/efeitos dos fármacos , Quinazolinas/uso terapêutico , Receptores de Interleucina-8B/antagonistas & inibidores , Triazóis/uso terapêutico , Adenocarcinoma/imunologia , Adenocarcinoma/metabolismo , Animais , Western Blotting , Proliferação de Células , Quimiotaxia , Meios de Cultivo Condicionados/farmacologia , Ensaio de Imunoadsorção Enzimática , Humanos , Técnicas Imunoenzimáticas , Técnicas In Vitro , Neoplasias Pulmonares/imunologia , Neoplasias Pulmonares/metabolismo , Melanoma/imunologia , Melanoma/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos SCID , RNA Mensageiro/genética , Receptores de Interleucina-8B/fisiologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Esferoides Celulares , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
4.
Mol Cancer Ther ; 18(5): 909-919, 2019 05.
Artigo em Inglês | MEDLINE | ID: mdl-30872381

RESUMO

Barasertib (AZD1152), a pro-drug of the highly potent and selective Aurora B kinase inhibitor AZD2811, showed promising clinical activity in relapsed/refractory diffuse large B-cell lymphoma (DLBCL) patients administered as a 4-day infusion. To improve potential therapeutic benefit of Aurora B kinase inhibition, a nanoparticle formulation of AZD2811 has been developed to address limitations of repeated intravenous infusion. One of the challenges with the use of nanoparticles for chronic treatment of tumors is optimizing dose and schedule required to enable repeat administration to sustain tumor growth inhibition. AZD2811 gives potent cell growth inhibition across a range of DLBCL cells lines in vitro In vivo, repeat administration of the AZD2811 nanoparticle gave antitumor activity at half the dose intensity of AZD1152. Compared with AZD1152, a single dose of AZD2811 nanoparticle gave less reduction in pHH3, but increased apoptosis and reduction of cells in G1 and G2-M, albeit at later time points, suggesting that duration and depth of target inhibition influence the nature of the tumor cell response to drug. Further exploration of the influence of dose and schedule on efficacy revealed that AZD2811 nanoparticle can be used flexibly with repeat administration of 25 mg/kg administered up to 7 days apart being sufficient to maintain equivalent tumor control. Timing of repeat administration could be varied with 50 mg/kg every 2 weeks controlling tumor control as effectively as 25 mg/kg every week. AZD2811 nanoparticle can be administered with very different doses and schedules to inhibit DLBCL tumor growth, although maximal tumor growth inhibition was achieved with the highest dose intensities.


Assuntos
Acetanilidas/farmacologia , Aurora Quinase B/genética , Linfoma Difuso de Grandes Células B/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Quinazolinas/farmacologia , Acetanilidas/química , Animais , Aurora Quinase B/antagonistas & inibidores , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Relação Dose-Resposta a Droga , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Linfoma Difuso de Grandes Células B/genética , Linfoma Difuso de Grandes Células B/patologia , Camundongos , Nanopartículas/química , Inibidores de Proteínas Quinases/química , Quinazolinas/química , Ensaios Antitumorais Modelo de Xenoenxerto
5.
Nat Commun ; 10(1): 2030, 2019 05 02.
Artigo em Inglês | MEDLINE | ID: mdl-31048689

RESUMO

Acquired resistance to MEK1/2 inhibitors (MEKi) arises through amplification of BRAFV600E or KRASG13D to reinstate ERK1/2 signalling. Here we show that BRAFV600E amplification and MEKi resistance are reversible following drug withdrawal. Cells with BRAFV600E amplification are addicted to MEKi to maintain a precise level of ERK1/2 signalling that is optimal for cell proliferation and survival, and tumour growth in vivo. Robust ERK1/2 activation following MEKi withdrawal drives a p57KIP2-dependent G1 cell cycle arrest and senescence or expression of NOXA and cell death, selecting against those cells with amplified BRAFV600E. p57KIP2 expression is required for loss of BRAFV600E amplification and reversal of MEKi resistance. Thus, BRAFV600E amplification confers a selective disadvantage during drug withdrawal, validating intermittent dosing to forestall resistance. In contrast, resistance driven by KRASG13D amplification is not reversible; rather ERK1/2 hyperactivation drives ZEB1-dependent epithelial-to-mesenchymal transition and chemoresistance, arguing strongly against the use of drug holidays in cases of KRASG13D amplification.


Assuntos
Antineoplásicos/farmacologia , Resistencia a Medicamentos Antineoplásicos/genética , Neoplasias/tratamento farmacológico , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas p21(ras)/genética , Antineoplásicos/uso terapêutico , Apoptose/efeitos dos fármacos , Apoptose/genética , Benzimidazóis/farmacologia , Benzimidazóis/uso terapêutico , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Transição Epitelial-Mesenquimal/genética , Feminino , Amplificação de Genes/efeitos dos fármacos , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , MAP Quinase Quinase 1/antagonistas & inibidores , MAP Quinase Quinase 2/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Sistema de Sinalização das MAP Quinases/genética , Masculino , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Neoplasias/genética , Inibidores de Proteínas Quinases/uso terapêutico , Suspensão de Tratamento , Homeobox 1 de Ligação a E-box em Dedo de Zinco/metabolismo
6.
Cancer Discov ; 9(2): 230-247, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30373918

RESUMO

Small cell lung cancer (SCLC) accounts for 15% of lung cancers and is almost always linked to inactivating RB1 and TP53 mutations. SCLC frequently responds, albeit briefly, to chemotherapy. The canonical function of the RB1 gene product RB1 is to repress the E2F transcription factor family. RB1 also plays both E2F-dependent and E2F-independent mitotic roles. We performed a synthetic lethal CRISPR/Cas9 screen in an RB1 -/- SCLC cell line that conditionally expresses RB1 to identify dependencies that are caused by RB1 loss and discovered that RB1 -/- SCLC cell lines are hyperdependent on multiple proteins linked to chromosomal segregation, including Aurora B kinase. Moreover, we show that an Aurora B kinase inhibitor is efficacious in multiple preclinical SCLC models at concentrations that are well tolerated in mice. These results suggest that RB1 loss is a predictive biomarker for sensitivity to Aurora B kinase inhibitors in SCLC and perhaps other RB1 -/- cancers. SIGNIFICANCE: SCLC is rarely associated with actionable protooncogene mutations. We did a CRISPR/Cas9-based screen that showed that RB1 -/- SCLC are hyperdependent on AURKB, likely because both genes control mitotic fidelity, and confirmed that Aurora B kinase inhibitors are efficacious against RB1 -/- SCLC tumors in mice at nontoxic doses.See related commentary by Dick and Li, p. 169.This article is highlighted in the In This Issue feature, p. 151.


Assuntos
Aurora Quinase B/metabolismo , Proliferação de Células , Genes Supressores de Tumor , Neoplasias Pulmonares/patologia , Mutação , Proteínas de Ligação a Retinoblastoma/metabolismo , Carcinoma de Pequenas Células do Pulmão/patologia , Ubiquitina-Proteína Ligases/metabolismo , Animais , Antineoplásicos/farmacologia , Apoptose , Aurora Quinase B/genética , Sistemas CRISPR-Cas , Segregação de Cromossomos , Resistencia a Medicamentos Antineoplásicos , Feminino , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Camundongos , Camundongos Nus , Proteínas de Ligação a Retinoblastoma/antagonistas & inibidores , Proteínas de Ligação a Retinoblastoma/genética , Transdução de Sinais , Carcinoma de Pequenas Células do Pulmão/tratamento farmacológico , Carcinoma de Pequenas Células do Pulmão/metabolismo , Células Tumorais Cultivadas , Ubiquitina-Proteína Ligases/antagonistas & inibidores , Ubiquitina-Proteína Ligases/genética , Ensaios Antitumorais Modelo de Xenoenxerto
7.
Mol Cancer Ther ; 16(6): 1031-1040, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28292940

RESUMO

Barasertib (AZD1152), a highly potent and selective aurora kinase B inhibitor, gave promising clinical activity in elderly acute myeloid leukemia (AML) patients. However, clinical utility was limited by the requirement for a 7-day infusion. Here we assessed the potential of a nanoparticle formulation of the selective Aurora kinase B inhibitor AZD2811 (formerly known as AZD1152-hQPA) in preclinical models of AML. When administered to HL-60 tumor xenografts at a single dose between 25 and 98.7 mg/kg, AZD2811 nanoparticle treatment delivered profound inhibition of tumor growth, exceeding the activity of AZD1152. The improved antitumor activity was associated with increased phospho-histone H3 inhibition, polyploidy, and tumor cell apoptosis. Moreover, AZD2811 nanoparticles increased antitumor activity when combined with cytosine arabinoside. By modifying dose of AZD2811 nanoparticle, therapeutic benefit in a range of preclinical models was further optimized. At high-dose, antitumor activity was seen in a range of models including the MOLM-13 disseminated model. At these higher doses, a transient reduction in bone marrow cellularity was observed demonstrating the potential for the formulation to target residual disease in the bone marrow, a key consideration when treating AML. Collectively, these data establish that AZD2811 nanoparticles have activity in preclinical models of AML. Targeting Aurora B kinase with AZD2811 nanoparticles is a novel approach to deliver a cell-cycle inhibitor in AML, and have potential to improve on the clinical activity seen with cell-cycle agents in this disease. Mol Cancer Ther; 16(6); 1031-40. ©2017 AACR.


Assuntos
Antineoplásicos/administração & dosagem , Aurora Quinase B/antagonistas & inibidores , Leucemia Mieloide Aguda/metabolismo , Leucemia Mieloide Aguda/patologia , Nanopartículas , Organofosfatos/administração & dosagem , Inibidores de Proteínas Quinases/administração & dosagem , Quinazolinas/administração & dosagem , Animais , Antineoplásicos/farmacocinética , Apoptose/efeitos dos fármacos , Medula Óssea/efeitos dos fármacos , Medula Óssea/metabolismo , Medula Óssea/patologia , Linhagem Celular Tumoral , Citarabina/farmacologia , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Quimioterapia Combinada , Feminino , Células HL-60 , Humanos , Leucemia Mieloide Aguda/tratamento farmacológico , Leucemia Mieloide Aguda/mortalidade , Camundongos , Organofosfatos/farmacocinética , Poliploidia , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/farmacocinética , Ratos , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
8.
Patient Educ Couns ; 62(3): 330-9, 2006 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-16860522

RESUMO

OBJECTIVE: For women who are experiencing intimate partner violence (IPV), making changes toward safety is often a gradual process. When providing counseling and support, health care providers may benefit from better understanding of where women are in their readiness to change. Our objective was to apply the transtheoretical model's stages of change to the experiences of women who experienced IPV and map their experiences of change as they moved toward increased safety. METHODS: A multi-disciplinary team designed a qualitative interview process with 20 women who had current or past histories of IPV in order to explore their experiences. RESULTS: The women in our study (1) moved through stages of readiness generally in a nonlinear fashion, with varying rates of progression between safe and nonsafe situations, (2) were able to identify a "turning-point" in their situations, (3) attempted multiple "action" steps and (4) were influenced by internal and external factors. CONCLUSIONS: Our study suggests that focusing on the transtheoretical model to develop stage-based interventions for IPV may not be the most appropriate given the nonsequential movement between stages and influence of external factors. PRACTICE IMPLICATIONS: The "change mapping" technique can be used as an educational and counseling tool with patients, as well as a training tool for health care providers.


Assuntos
Adaptação Psicológica , Mulheres Maltratadas/psicologia , Comportamentos Relacionados com a Saúde , Modelos Psicológicos , Aceitação pelo Paciente de Cuidados de Saúde/psicologia , Maus-Tratos Conjugais/psicologia , Adulto , Mulheres Maltratadas/educação , Aconselhamento/organização & administração , Escolaridade , Feminino , Conhecimentos, Atitudes e Prática em Saúde , Humanos , Pessoa de Meia-Idade , Motivação , Narração , Educação de Pacientes como Assunto/organização & administração , Pennsylvania , Pesquisa Qualitativa , Gestão da Segurança , Autocuidado/métodos , Autocuidado/psicologia , Fatores Socioeconômicos , Maus-Tratos Conjugais/prevenção & controle , Inquéritos e Questionários , Fatores de Tempo
9.
Cancer Res ; 62(20): 5749-54, 2002 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-12384534

RESUMO

The epidermal growth factor receptor (EGFR) is a promising target for anticancer therapy because of its role in tumor growth, metastasis and angiogenesis, and tumor resistance to chemotherapy and radiotherapy. We have developed a low-molecular-weight EGFR tyrosine kinase inhibitor (EGFR-TKI), ZD1839 (Iressa(2) ). ZD1839, a substituted anilinoquinazoline, is a potent EGFR-TKI (IC(50) = 0.033 micro M) that selectively inhibits EGF-stimulated tumor cell growth (IC(50) = 0.054 micro M) and that blocks EGF-stimulated EGFR autophosphorylation in tumor cells. In studies with mice bearing a range of human tumor-derived xenografts, ZD1839 given p.o. once a day inhibited tumor growth in a dose-dependent manner. The level of expression of EGFR did not determine xenograft tumor sensitivity to ZD1839. Long-term ZD1839 (>3 months) treatment of mice bearing A431 xenografts was well tolerated, and ZD1839 completely inhibited tumor growth and induced regression of established tumors. No drug-resistant tumors appeared during ZD1839 treatment, but some tumors regrew after drug withdrawal. These studies indicate the potential utility of ZD1839 in the treatment of many human tumors and indicate that continuous once-a-day p.o. dosing might be a suitable therapeutic regimen.


Assuntos
Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Quinazolinas/farmacologia , Administração Oral , Animais , Divisão Celular/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Receptores ErbB/metabolismo , Receptores ErbB/fisiologia , Feminino , Gefitinibe , Inibidores do Crescimento/farmacologia , Humanos , Camundongos , Camundongos Nus , Fosforilação/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Dementia (London) ; 15(5): 958-75, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25187482

RESUMO

BACKGROUND: End of life decisions for people with advanced dementia are reported as often being difficult for families as they attempt to make appropriate and justified decisions. AIM: To explore the experiences of advance care planning amongst family caregivers of people with advanced dementia. DESIGN: Qualitative research including a series of single cases (close family relatives). METHODS: A purposive sample of 12 family caregivers within a specialist dementia unit was interviewed about their experiences of advance care planning between August 2009 and February 2010. RESULTS/FINDINGS: Family caregivers need encouragement to ask the right questions during advance care planning to discuss the appropriateness of nursing and medical interventions at the end of life. CONCLUSIONS: Advance care planning can be facilitated with the family caregiver in the context of everyday practice within the nursing home environment for older people with dementia.


Assuntos
Planejamento Antecipado de Cuidados , Cuidadores/psicologia , Demência/psicologia , Assistência Terminal/psicologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Tomada de Decisões , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Pesquisa Qualitativa
11.
Mol Cancer Ther ; 15(10): 2378-2387, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-27439477

RESUMO

Osimertinib (AZD9291) is a potent, selective, irreversible inhibitor of EGFR-sensitizing (exon 19 and L858R) and T790M-resistant mutation. In vivo, in the mouse, it is metabolized to an active des-methyl metabolite, AZ5104. To understand the therapeutic potential in patients, this study aimed to assess the relationship between osimertinib pharmacokinetics, the pharmacokinetics of the active metabolite, the pharmacodynamics of phosphorylated EGFR reduction, and efficacy in mouse xenograft models of EGFR-driven cancers, including two NSCLC lines. Osimertinib was dosed in xenografted models of EGFR-driven cancers. In one set of experiments, changes in phosphorylated EGFR were measured to confirm target engagement. In a second set of efficacy studies, the resulting changes in tumor volume over time after repeat dosing of osimertinib were observed. To account for the contributions of both molecules, a mathematical modeling approach was taken to integrate the resulting datasets. The model was able to describe the pharmacokinetics, pharmacodynamics, and efficacy in A431, PC9, and NCI-H1975 xenografts, with the differences in sensitivity described by the varying potency against wild-type, sensitizing, and T790M-mutant EGFR and the phosphorylated EGFR reduction required to reduce tumor volume. It was inferred that recovery of pEGFR is slower after chronic dosing due to reduced resynthesis. It was predicted and further demonstrated that although inhibition is irreversible, the resynthesis of EGFR is such that infrequent intermittent dosing is not as efficacious as once daily dosing. Mol Cancer Ther; 15(10); 2378-87. ©2016 AACR.


Assuntos
Antineoplásicos/farmacocinética , Receptores ErbB/antagonistas & inibidores , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacocinética , Acrilamidas , Algoritmos , Compostos de Anilina , Animais , Antineoplásicos/química , Linhagem Celular Tumoral , Modelos Animais de Doenças , Receptores ErbB/química , Humanos , Camundongos , Modelos Biológicos , Piperazinas/química , Inibidores de Proteínas Quinases/química , Ensaios Antitumorais Modelo de Xenoenxerto
12.
Oncotarget ; 7(52): 86313-86325, 2016 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-27861144

RESUMO

Metabolic plasticity is an emerging hallmark of cancer, and increased glycolysis is often observed in transformed cells. Small molecule inhibitors that target driver oncogenes can potentially inhibit the glycolytic pathway. Osimertinib (AZD9291) is a novel EGFR tyrosine kinase inhibitor (TKI) that is potent and selective for sensitising (EGFRm) and T790M resistance mutations. Clinical studies have shown osimertinib to be efficacious in patients with EGFRm/ T790M advanced NSCLC who have progressed after EGFR-TKI treatment. However experience with targeted therapies suggests that acquired resistance may emerge. Thus there is a need to characterize resistance mechanisms and to devise ways to prevent, delay or overcome osimertinib resistance. We show here that osimertinib suppresses glycolysis in parental EGFR-mutant lung adenocarcinoma lines, but has not in osimertinib-resistant cell lines. Critically, we show osimertinib treatment induces a strict dependence on mitochondrial oxidative phosphorylation (OxPhos), as OxPhos inhibitors significantly delay the long-term development of osimertinib resistance in osimertinib-sensitive lines. Accordingly, growth conditions which promote a less glycolytic phenotype confer a degree of osimertinib resistance. Our data support a model in which the combination of osimertinib and OxPhos inhibitors can delay or prevent resistance in osimertinib-naïve tumour cells, and represents a novel strategy that warrants further pre-clinical investigation.


Assuntos
Adenocarcinoma/tratamento farmacológico , Antineoplásicos/farmacologia , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Fosforilação Oxidativa/efeitos dos fármacos , Piperazinas/farmacologia , Inibidores de Proteínas Quinases/farmacologia , Acrilamidas , Adenocarcinoma/patologia , Adenocarcinoma de Pulmão , Compostos de Anilina , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB/genética , Glicólise/efeitos dos fármacos , Humanos , Neoplasias Pulmonares/patologia
13.
J Control Release ; 229: 106-119, 2016 05 10.
Artigo em Inglês | MEDLINE | ID: mdl-27001894

RESUMO

The present studies were aimed at formulating AZD2811-loaded polylactic acid-polyethylene glycol (PLA-PEG) nanoparticles with adjustable release rates without altering the chemical structures of the polymer or active pharmaceutical ingredient (API). This was accomplished through the use of a hydrophobic ion pairing approach. A series of AZD2811-containing nanoparticles with a variety of hydrophobic counterions including oleic acid, 1-hydroxy-2-naphthoic acid, cholic acid, deoxycholic acid, dioctylsulfosuccinic acid, and pamoic acid is described. The hydrophobicity of AZD2811 was increased through formation of ion pairs with these hydrophobic counterions, producing nanoparticles with exceptionally high drug loading-up to five fold higher encapsulation efficiency and drug loading compared to nanoparticles made without hydrophobic ion pairs. Furthermore, the rate at which the drug was released from the nanoparticles could be controlled by employing counterions with various hydrophobicities and structures, resulting in release half-lives ranging from about 2 to 120h using the same polymer, nanoparticle size, and nanoemulsion process. Process recipe variables affecting drug load and release rate were identified, including pH and molarity of quench buffer. Ion pair formation between AZD2811 and pamoic acid as a model counterion was investigated using solubility enhancement as well as nuclear magnetic resonance spectroscopy to demonstrate solution-state interactions. Further evidence for an ion pairing mechanism of controlled release was provided through the measurement of API and counterion release profiles using high-performance liquid chromatography, which had stoichiometric relationships. Finally, Raman spectra of an AZD2811-pamoate salt compared well with those of the formulated nanoparticles, while single components (AZD2811, pamoic acid) alone did not. A library of AZD2811 batches was created for analytical and preclinical characterization. Dramatically improved preclinical efficacy and tolerability data were generated for the pamoic acid lead formulation, which has been selected for evaluation in a Phase 1 clinical trial (ClinicalTrials.gov Identifier NCT 02579226). This work clearly demonstrates the importance of assessing a wide range of drug release rates during formulation screening as a critical step for new drug product development, and how utilizing hydrophobic ion pairing enabled this promising nanoparticle formulation to proceed into clinical development.


Assuntos
Acetanilidas/administração & dosagem , Antineoplásicos , Sistemas de Liberação de Medicamentos , Nanopartículas , Organofosfatos , Pró-Fármacos , Quinazolinas/administração & dosagem , Acetanilidas/química , Acetanilidas/farmacocinética , Acetanilidas/uso terapêutico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos/química , Antineoplásicos/farmacocinética , Antineoplásicos/uso terapêutico , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Linhagem Celular Tumoral , Ácido Cólico/química , Ácido Desoxicólico/química , Ácido Dioctil Sulfossuccínico/química , Humanos , Interações Hidrofóbicas e Hidrofílicas , Masculino , Camundongos Nus , Nanopartículas/administração & dosagem , Nanopartículas/química , Nanopartículas/uso terapêutico , Naftóis/química , Neoplasias/tratamento farmacológico , Neoplasias/patologia , Organofosfatos/administração & dosagem , Organofosfatos/química , Organofosfatos/farmacocinética , Organofosfatos/uso terapêutico , Polietilenoglicóis/química , Pró-Fármacos/administração & dosagem , Pró-Fármacos/química , Pró-Fármacos/farmacocinética , Pró-Fármacos/uso terapêutico , Quinazolinas/química , Quinazolinas/farmacocinética , Quinazolinas/uso terapêutico , Ratos Nus , Ratos Wistar , Carga Tumoral/efeitos dos fármacos
14.
ACS Med Chem Lett ; 7(5): 514-9, 2016 May 12.
Artigo em Inglês | MEDLINE | ID: mdl-27190603

RESUMO

A novel series of covalent inhibitors of EGFR (epidermal growth factor receptor) kinase was discovered through a combination of subset screening and structure-based design. These compounds preferentially inhibit mutant forms of EGFR (activating mutant and T790M mutant) over wild-type EGFR in cellular assays measuring EGFR autophosphorylation and proliferation, suggesting an improved therapeutic index in non-small cell lung cancer patients would be achievable relative to established EGFR inhibitors. We describe our design approaches, resulting in the identification of the lead compound 5, and our efforts to develop an understanding of the structure-activity relationships within this series. In addition, strategies to overcome challenges around metabolic stability and aqueous solubility are discussed. Despite limitations in its physical properties, 5 is orally bioavailable in mice and demonstrates pronounced antitumor activity in in vivo models of mutant EGFR-driven cancers.

15.
Sci Transl Med ; 8(325): 325ra17, 2016 Feb 10.
Artigo em Inglês | MEDLINE | ID: mdl-26865565

RESUMO

Efforts to apply nanotechnology in cancer have focused almost exclusively on the delivery of cytotoxic drugs to improve therapeutic index. There has been little consideration of molecularly targeted agents, in particular kinase inhibitors, which can also present considerable therapeutic index limitations. We describe the development of Accurin polymeric nanoparticles that encapsulate the clinical candidate AZD2811, an Aurora B kinase inhibitor, using an ion pairing approach. Accurins increase biodistribution to tumor sites and provide extended release of encapsulated drug payloads. AZD2811 nanoparticles containing pharmaceutically acceptable organic acids as ion pairing agents displayed continuous drug release for more than 1 week in vitro and a corresponding extended pharmacodynamic reduction of tumor phosphorylated histone H3 levels in vivo for up to 96 hours after a single administration. A specific AZD2811 nanoparticle formulation profile showed accumulation and retention in tumors with minimal impact on bone marrow pathology, and resulted in lower toxicity and increased efficacy in multiple tumor models at half the dose intensity of AZD1152, a water-soluble prodrug of AZD2811. These studies demonstrate that AZD2811 can be formulated in nanoparticles using ion pairing agents to give improved efficacy and tolerability in preclinical models with less frequent dosing. Accurins specifically, and nanotechnology in general, can increase the therapeutic index of molecularly targeted agents, including kinase inhibitors targeting cell cycle and oncogenic signal transduction pathways, which have to date proved toxic in humans.


Assuntos
Aurora Quinases/antagonistas & inibidores , Nanopartículas/química , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Animais , Aurora Quinases/metabolismo , Medula Óssea/efeitos dos fármacos , Medula Óssea/patologia , Linhagem Celular Tumoral , Liberação Controlada de Fármacos , Feminino , Humanos , Masculino , Espectrometria de Massas , Camundongos , Camundongos SCID , Organofosfatos/química , Organofosfatos/farmacocinética , Organofosfatos/farmacologia , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacocinética , Quinazolinas/química , Quinazolinas/farmacocinética , Quinazolinas/farmacologia , Ratos Nus , Resultado do Tratamento , Ensaios Antitumorais Modelo de Xenoenxerto
16.
J Med Chem ; 48(2): 392-402, 2005 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-15658853

RESUMO

Thymidine phosphorylase (TP) is an important target enzyme for cancer chemotherapy because it is expressed at high levels in the hypoxic regions of many tumors and inhibitors of TP have been shown in animal model studies to inhibit angiogenesis and metastasis, and to promote tumor cell apoptosis. The 5-halo-6-[(2'-aminoimidazol-1'-yl)methyl]uracils (3, X = Cl, Br) are very potent inhibitors of E. coli and human TP with IC(50) values of approximately 20 nM when the enzyme concentration is approximately 40 nM. Their 4'-aminoimidazol-1'-yl analogues (4, X = Cl, Br) are >350-fold less active with IC(50) values of approximately 7 microM. The 5-unsubstituted analogues (3 and 4, X = H) were both less active than their 5-halo derivatives. Determination of pK(a) values and molecular modeling studies of these compounds in the active site of human TP was used to rationalize their activities. The finding that 3, X = Br has a poor pharmacokinetic (PK) profile in mice, coupled with the desire for tumor selectivity, led us to design prodrugs. The corresponding 2'-nitroimidazol-1'-ylmethyluracils (5, X = Cl, Br) are >1000-fold less active (IC(50) 22-24 microM) than their 2'-amino analogues and are reduced to the 2'-amino inhibitors (3, X = Cl, Br) by xanthine oxidase (XO). As XO is also highly expressed in many tumors, the 2'-nitro prodrugs have the potential to selectively deliver the potent 2'-aminoimidazol-1'-yl TP inhibitors into hypoxic solid tumors.


Assuntos
Antineoplásicos/síntese química , Imidazóis/síntese química , Pró-Fármacos/síntese química , Timidina Fosforilase/antagonistas & inibidores , Uracila/análogos & derivados , Uracila/síntese química , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Sítios de Ligação , Disponibilidade Biológica , Escherichia coli/química , Humanos , Imidazóis/química , Imidazóis/farmacologia , Camundongos , Modelos Moleculares , Nitroimidazóis/síntese química , Nitroimidazóis/química , Nitroimidazóis/farmacologia , Oxirredução , Pró-Fármacos/química , Pró-Fármacos/farmacologia , Relação Estrutura-Atividade , Uracila/farmacologia
17.
Int J Radiat Oncol Biol Phys ; 54(5): 1497-502, 2002 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-12459377

RESUMO

PURPOSE: The aim of these studies was to evaluate factors that contribute to the selectivity of the novel vascular targeting agent ZD6126. METHODS: Human umbilical vein endothelial cells (HUVECs) were treated with ZD6126 phenol, and effects on morphology, detachment, and cytotoxicity (sulforhodamine-B dye incorporation) were determined. Hras5-transformed mouse 3T3 fibroblasts were implanted s.c. in athymic nude rats, and effects on the tumor were assessed after either i.v. bolus or 24-h minipump infusion of ZD6126. RESULTS: In vitro, ZD6126 phenol ( approximately 0.1 microm) rapidly (<40 min) destabilized the tubulin cytoskeleton of proliferating endothelial cells, resulting in cell shape change ("rounding up") and cell detachment at noncytotoxic drug concentrations. In vivo, in rats, an i.v. bolus dose of ZD6126 (20 mg/kg) was rapidly broken down to ZD6126 phenol, which has a short plasma elimination half-life ( approximately 1 h). Peak plasma levels of ZD6126 phenol were well above the level required to induce HUVEC morphology changes in vitro, but cytotoxic concentrations were not maintained. A single i.v. bolus dose (50 and 20 mg/kg) of ZD6126 was well tolerated and resulted in extensive central tumor necrosis in the Hras5 model. Administration of ZD6126 using a 24-h s.c. minipump resulted in decreased ( approximately 30-fold) peak plasma levels, but maintained cytotoxic drug levels over 24 h. Infusion of 50 mg/kg ZD6126 over 24 h was not tolerated. Infusion of 20 mg/kg ZD6126 resulted in increased toxicity compared with the i.v. bolus doses of ZD6126 and did not result in any increased tumor necrosis after 24 h. CONCLUSION: ZD6126 phenol induces rapid morphologic changes in HUVECs at noncytotoxic drug levels. These rapid morphologic effects combined with the rapid elimination of ZD6126 phenol contribute to the selective effects of ZD6126 on tumor vasculature at well-tolerated doses.


Assuntos
Inibidores da Angiogênese/uso terapêutico , Endotélio Vascular/metabolismo , Neovascularização Patológica , Compostos Organofosforados/uso terapêutico , Células 3T3 , Animais , Divisão Celular , Linhagem Celular Transformada , Células Cultivadas , Relação Dose-Resposta a Droga , Endotélio Vascular/citologia , Fibroblastos/metabolismo , Humanos , Camundongos , Necrose , Transplante de Neoplasias , Ratos , Ratos Nus , Fatores de Tempo , Veias Umbilicais/citologia
18.
Cancer Discov ; 4(9): 1046-61, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24893891

RESUMO

UNLABELLED: First-generation EGFR tyrosine kinase inhibitors (EGFR TKI) provide significant clinical benefit in patients with advanced EGFR-mutant (EGFRm(+)) non-small cell lung cancer (NSCLC). Patients ultimately develop disease progression, often driven by acquisition of a second T790M EGFR TKI resistance mutation. AZD9291 is a novel oral, potent, and selective third-generation irreversible inhibitor of both EGFRm(+) sensitizing and T790M resistance mutants that spares wild-type EGFR. This mono-anilino-pyrimidine compound is structurally distinct from other third-generation EGFR TKIs and offers a pharmacologically differentiated profile from earlier generation EGFR TKIs. Preclinically, the drug potently inhibits signaling pathways and cellular growth in both EGFRm(+) and EGFRm(+)/T790M(+) mutant cell lines in vitro, with lower activity against wild-type EGFR lines, translating into profound and sustained tumor regression in EGFR-mutant tumor xenograft and transgenic models. The treatment of 2 patients with advanced EGFRm(+) T790M(+) NSCLC is described as proof of principle. SIGNIFICANCE: We report the development of a novel structurally distinct third-generation EGFR TKI, AZD9291, that irreversibly and selectively targets both sensitizing and resistant T790M(+) mutant EGFR while harboring less activity toward wild-type EGFR. AZD9291 is showing promising responses in a phase I trial even at the first-dose level, with first published clinical proof-of-principle validation being presented.


Assuntos
Acrilamidas/uso terapêutico , Compostos de Anilina/uso terapêutico , Antineoplásicos/uso terapêutico , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/genética , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/genética , Mutação , Inibidores de Proteínas Quinases/uso terapêutico , Acrilamidas/química , Acrilamidas/farmacologia , Compostos de Anilina/química , Compostos de Anilina/farmacologia , Animais , Antineoplásicos/química , Antineoplásicos/farmacologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Modelos Animais de Doenças , Receptores ErbB/química , Feminino , Genes erbB-2 , Humanos , Neoplasias Pulmonares/patologia , Masculino , Pessoa de Meia-Idade , Modelos Moleculares , Conformação Molecular , Fosforilação , Ligação Proteica , Inibidores de Proteínas Quinases/química , Inibidores de Proteínas Quinases/farmacologia , Transdução de Sinais/efeitos dos fármacos , Resultado do Tratamento , Carga Tumoral/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
19.
J Med Chem ; 57(20): 8249-67, 2014 Oct 23.
Artigo em Inglês | MEDLINE | ID: mdl-25271963

RESUMO

Epidermal growth factor receptor (EGFR) inhibitors have been used clinically in the treatment of non-small-cell lung cancer (NSCLC) patients harboring sensitizing (or activating) mutations for a number of years. Despite encouraging clinical efficacy with these agents, in many patients resistance develops leading to disease progression. In most cases, this resistance is in the form of the T790M mutation. In addition, EGFR wild type receptor inhibition inherent with these agents can lead to dose limiting toxicities of rash and diarrhea. We describe herein the evolution of an early, mutant selective lead to the clinical candidate AZD9291, an irreversible inhibitor of both EGFR sensitizing (EGFRm+) and T790M resistance mutations with selectivity over the wild type form of the receptor. Following observations of significant tumor inhibition in preclinical models, the clinical candidate was administered clinically to patients with T790M positive EGFR-TKI resistant NSCLC and early efficacy has been observed, accompanied by an encouraging safety profile.


Assuntos
Acrilamidas/farmacologia , Compostos de Anilina/farmacologia , Antineoplásicos/farmacologia , Carcinoma Pulmonar de Células não Pequenas/tratamento farmacológico , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/antagonistas & inibidores , Neoplasias Pulmonares/tratamento farmacológico , Animais , Antineoplásicos/química , Antineoplásicos/farmacocinética , Carcinoma Pulmonar de Células não Pequenas/genética , Técnicas de Química Sintética , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Receptores ErbB/genética , Feminino , Humanos , Concentração Inibidora 50 , Neoplasias Pulmonares/genética , Masculino , Camundongos , Pessoa de Meia-Idade , Mutação , Inibidores de Proteínas Quinases/síntese química , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Ratos Endogâmicos , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Med Chem ; 56(17): 7025-48, 2013 Sep 12.
Artigo em Inglês | MEDLINE | ID: mdl-23930994

RESUMO

A novel series of small-molecule inhibitors has been developed to target the double mutant form of the epidermal growth factor receptor (EGFR) tyrosine kinase, which is resistant to treatment with gefitinib and erlotinib. Our reported compounds also show selectivity over wild-type EGFR. Guided by molecular modeling, this series was evolved to target a cysteine residue in the ATP binding site via covalent bond formation and demonstrates high levels of activity in cellular models of the double mutant form of EGFR. In addition, these compounds show significant activity against the activating mutations, which gefitinib and erlotinib target and inhibition of which gives rise to their observed clinical efficacy. A glutathione (GSH)-based assay was used to measure thiol reactivity toward the electrophilic functionality of the inhibitor series, enabling both the identification of a suitable reactivity window for their potency and the development of a reactivity quantitative structure-property relationship (QSPR) to support design.


Assuntos
Receptores ErbB/antagonistas & inibidores , Receptores ErbB/química , Receptores ErbB/genética , Modelos Moleculares , Mutação , Relação Estrutura-Atividade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA