Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Drug Resist Updat ; 74: 101078, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38503142

RESUMO

AIMS: Human epidermal growth factor receptor 2 (HER2) is an oncogenic receptor tyrosine kinase amplified in approximately 20% of breast cancer (BC). HER2-targeted therapies are the linchpin of treating HER2-positive BC. However, drug resistance is common, and the main resistance mechanism is unknown. We tested the hypothesis that drug resistance results mainly from inadequate or lack of inhibition of HER2 and its family member epidermal growth factor receptor (EGFR). METHODS: We used clinically relevant cell and tumor models to assess the impact of targeted degradation of HER2 and EGFR on trastuzumab resistance. Trastuzumab is the most common clinically used HER2 inhibitor. Targeted degradation of HER2 and EGFR was achieved using recombinant human protein PEPDG278D, which binds to the extracellular domains of the receptors. siRNA knockdown was used to assess the relative importance of EGFR and HER2 in trastuzumab resistance. RESULTS: Both HER2 and EGFR are overexpressed in all trastuzumab-resistant HER2-positive BC cell and tumor models and that all trastuzumab-resistant models are highly vulnerable to targeted degradation of HER2 and EGFR. Degradation of HER2 and EGFR induced by PEPDG278D causes extensive inhibition of oncogenic signaling in trastuzumab-resistant HER2-positive BC cells. This is accompanied by strong growth inhibition of cultured cells, orthotopic patient-derived xenografts, and metastatic lesions in the brain and lung of trastuzumab-resistant HER2-positive BC. siRNA knockdown indicates that eliminating both HER2 and EGFR is necessary to maximize therapeutic outcome. CONCLUSIONS: This study unravels the therapeutic vulnerability of trastuzumab-resistant HER2-positive BC and shows that an agent that targets the degradation of both HER2 and EGFR is highly effective in overcoming drug resistance in this disease. The findings provide new insights and innovations for advancing treatment of drug-resistant HER2-positive breast cancer that remains an unmet problem.


Assuntos
Neoplasias da Mama , Resistencia a Medicamentos Antineoplásicos , Receptores ErbB , Receptor ErbB-2 , Transdução de Sinais , Trastuzumab , Humanos , Neoplasias da Mama/tratamento farmacológico , Neoplasias da Mama/patologia , Neoplasias da Mama/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Feminino , Trastuzumab/farmacologia , Trastuzumab/uso terapêutico , Receptor ErbB-2/metabolismo , Receptor ErbB-2/antagonistas & inibidores , Animais , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Camundongos , Transdução de Sinais/efeitos dos fármacos , Linhagem Celular Tumoral , Ensaios Antitumorais Modelo de Xenoenxerto , Proteólise/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos
2.
Indoor Air ; 31(2): 587-601, 2021 03.
Artigo em Inglês | MEDLINE | ID: mdl-32870542

RESUMO

Human activity is known to leave significant effects on indoor airflow patterns. These patterns are carefully designed for many facilities such as cleanrooms, pharmaceutical settings, and healthcare environments, where human-induced wakes contribute to the transport of contaminants. Therefore, the knowledge about these wakes as it relates to indoor air quality is critical. As a result, a series of experiments were conducted in a controlled chamber to study the three-dimensional effects of true human walking on airflow. Experiments were designed to capture the effect of human walking under three different flow conditions, and for two different walking schemes. The results show that the effect of walking on the airflow is not negligible and can sustain up to 10 seconds after the moving body has passed. Walking on a straight line creates significant change in the velocity normal to the walking path and vertical to the plane of walking movement. These changes were detectable till 1.0 m away from the walking track. Also, the similarity between airflow patterns of walking once and twice illustrated a promising opportunity of predicting the flow patterns of random walk from a set of base cases.


Assuntos
Movimentos do Ar , Poluição do Ar em Ambientes Fechados , Simulação por Computador , Humanos , Movimento , Fenômenos Fisiológicos Respiratórios , Caminhada
3.
JAMA ; 323(13): 1257-1265, 2020 04 07.
Artigo em Inglês | MEDLINE | ID: mdl-32078683

RESUMO

Importance: Intravenous thrombolysis with tenecteplase improves reperfusion prior to endovascular thrombectomy for ischemic stroke compared with alteplase. Objective: To determine whether 0.40 mg/kg of tenecteplase safely improves reperfusion before endovascular thrombectomy vs 0.25 mg/kg of tenecteplase in patients with large vessel occlusion ischemic stroke. Design, Setting, and Participants: Randomized clinical trial at 27 hospitals in Australia and 1 in New Zealand using open-label treatment and blinded assessment of radiological and clinical outcomes. Patients were enrolled from December 2017 to July 2019 with follow-up until October 2019. Adult patients (N = 300) with ischemic stroke due to occlusion of the intracranial internal carotid, \basilar, or middle cerebral artery were included less than 4.5 hours after symptom onset using standard intravenous thrombolysis eligibility criteria. Interventions: Open-label tenecteplase at 0.40 mg/kg (maximum, 40 mg; n = 150) or 0.25 mg/kg (maximum, 25 mg; n = 150) given as a bolus before endovascular thrombectomy. Main Outcomes and Measures: The primary outcome was reperfusion of greater than 50% of the involved ischemic territory prior to thrombectomy, assessed by consensus of 2 blinded neuroradiologists. Prespecified secondary outcomes were level of disability at day 90 (modified Rankin Scale [mRS] score; range, 0-6); mRS score of 0 to 1 (freedom from disability) or no change from baseline at 90 days; mRS score of 0 to 2 (functional independence) or no change from baseline at 90 days; substantial neurological improvement at 3 days; symptomatic intracranial hemorrhage within 36 hours; and all-cause death. Results: All 300 patients who were randomized (mean age, 72.7 years; 141 [47%] women) completed the trial. The number of participants with greater than 50% reperfusion of the previously occluded vascular territory was 29 of 150 (19.3%) in the 0.40 mg/kg group vs 29 of 150 (19.3%) in the 0.25 mg/kg group (unadjusted risk difference, 0.0% [95% CI, -8.9% to -8.9%]; adjusted risk ratio, 1.03 [95% CI, 0.66-1.61]; P = .89). Among the 6 secondary outcomes, there were no significant differences in any of the 4 functional outcomes between the 0.40 mg/kg and 0.25 mg/kg groups nor in all-cause deaths (26 [17%] vs 22 [15%]; unadjusted risk difference, 2.7% [95% CI, -5.6% to 11.0%]) or symptomatic intracranial hemorrhage (7 [4.7%] vs 2 [1.3%]; unadjusted risk difference, 3.3% [95% CI, -0.5% to 7.2%]). Conclusions and Relevance: Among patients with large vessel occlusion ischemic stroke, a dose of 0.40 mg/kg, compared with 0.25 mg/kg, of tenecteplase did not significantly improve cerebral reperfusion prior to endovascular thrombectomy. The findings suggest that the 0.40-mg/kg dose of tenecteplase does not confer an advantage over the 0.25-mg/kg dose in patients with large vessel occlusion ischemic stroke in whom endovascular thrombectomy is planned. Trial Registration: ClinicalTrials.gov Identifier: NCT03340493.


Assuntos
Fibrinolíticos/administração & dosagem , Reperfusão/métodos , Acidente Vascular Cerebral/tratamento farmacológico , Tenecteplase/administração & dosagem , Trombectomia , Idoso , Idoso de 80 Anos ou mais , Isquemia Encefálica/tratamento farmacológico , Relação Dose-Resposta a Droga , Feminino , Fibrinolíticos/efeitos adversos , Humanos , Masculino , Pessoa de Meia-Idade , Acidente Vascular Cerebral/cirurgia , Tenecteplase/efeitos adversos , Resultado do Tratamento
4.
Int J Mol Sci ; 19(11)2018 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-30380599

RESUMO

Durable response, inherent or acquired resistance, and dose-limiting toxicities continue to represent major barriers in the treatment of patients with advanced clear-cell renal cell carcinoma (ccRCC). The majority of ccRCC tumors are characterized by the loss of Von Hippel⁻Lindau tumor suppressor gene function, a stable expression of hypoxia-inducible factors 1α and 2α (HIFs), an altered expression of tumor-specific oncogenic microRNAs (miRNAs), a clear cytoplasm with dense lipid content, and overexpression of thymidine phosphorylase. The aim of this manuscript was to confirm that the downregulation of specific drug-resistant biomarkers deregulated in tumor cells by a defined dose and schedule of methylselenocysteine (MSC) or seleno-l-methionine (SLM) sensitizes tumor cells to mechanism-based drug combination. The inhibition of HIFs by selenium was necessary for optimal therapeutic benefit. Durable responses were achieved only when MSC was combined with sunitinib (a vascular endothelial growth factor receptor (VEGFR)-targeted biologic), topotecan (a topoisomerase 1 poison and HIF synthesis inhibitor), and S-1 (a 5-fluorouracil prodrug). The documented synergy was selenium dose- and schedule-dependent and associated with enhanced prolyl hydroxylase-dependent HIF degradation, stabilization of tumor vasculature, downregulation of 28 oncogenic miRNAs, as well as the upregulation of 12 tumor suppressor miRNAs. The preclinical results generated provided the rationale for the development of phase 1/2 clinical trials of SLM in sequential combination with axitinib in ccRCC patients refractory to standard therapies.


Assuntos
Antineoplásicos/uso terapêutico , Fatores de Transcrição Hélice-Alça-Hélice Básicos/genética , Carcinoma de Células Renais/tratamento farmacológico , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Neoplasias Renais/tratamento farmacológico , MicroRNAs/genética , Selenocisteína/análogos & derivados , Selenometionina/uso terapêutico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Carcinoma de Células Renais/irrigação sanguínea , Carcinoma de Células Renais/genética , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Feminino , Fluoruracila/uso terapêutico , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Neoplasias Renais/irrigação sanguínea , Neoplasias Renais/genética , Neoplasias Renais/patologia , Camundongos Nus , Selenocisteína/uso terapêutico , Topotecan/uso terapêutico
5.
IEEE J Transl Eng Health Med ; 12: 291-297, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38410180

RESUMO

OBJECTIVE: A change in handwriting is an early sign of Parkinson's disease (PD). However, significant inter-person differences in handwriting make it difficult to identify pathological handwriting, especially in the early stages. This paper reports the testing of NeuroDiag, a software-based medical device, for the automated detection of PD using handwriting patterns. NeuroDiag is designed to direct the user to perform six drawing and writing tasks, and the recordings are then uploaded onto a server for analysis. Kinematic information and pen pressure of handwriting are extracted and used as baseline parameters. NeuroDiag was trained based on 26 PD patients in the early stage of the disease and 26 matching controls. METHODS: Twenty-three people with PD (PPD) in their early stage of the disease, 25 age-matched healthy controls (AMC), and 7 young healthy controls were recruited for this study. Under the supervision of a consultant neurologist or their nurse, the participants used NeuroDiag. The reports were generated in real-time and tabulated by an independent observer. RESULTS: The participants were able to use NeuroDiag without assistance. The handwriting data was successfully uploaded to the server where the report was automatically generated in real-time. There were significant differences in the writing speed between PPD and AMC (P<0.001). NeuroDiag showed 86.96% sensitivity and 76.92% specificity in differentiating PPD from those without PD. CONCLUSION: In this work, we tested the reliability of NeuroDiag in differentiating between PPD and AMC for real-time applications. The results show that NeuroDiag has the potential to be used to assist neurologists and for telehealth applications. Clinical and Translational Impact Statement - This pre-clinical study shows the feasibility of developing a community-wide screening program for Parkinson's disease using automated handwriting analysis software, NeuroDiag.


Assuntos
Doença de Parkinson , Humanos , Doença de Parkinson/diagnóstico , Reprodutibilidade dos Testes , Escrita Manual , Software , Fenômenos Biomecânicos
6.
Carcinogenesis ; 34(11): 2593-9, 2013 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-23946495

RESUMO

Allyl isothiocyanate (AITC) occurs in cruciferous vegetables that are commonly consumed by humans and has been shown to inhibit urinary bladder cancer growth and progression in previous preclinical studies. However, AITC does not significantly modulate cyclooxygenase-2 (Cox-2), whose oncogenic activity has been well documented in bladder cancer and other cancers. Celecoxib is a selective Cox-2 inhibitor and has been widely used for treatment of several diseases. Celecoxib has also been evaluated in bladder cancer patients, but its efficacy against bladder cancer as a single agent remains unclear. In a syngeneic rat model of orthotopic bladder cancer, treatment of the animals with the combination of AITC and celecoxib at low dose levels (AITC at 1 mg/kg and celecoxib at 10 mg/kg) led to increased or perhaps synergistic inhibition of bladder cancer growth and muscle invasion, compared with each agent used alone. The combination regime was also more effective than each single agent in inhibiting microvessel formation and stimulating microvessel maturation in the tumor tissues. The anticancer efficacy of the combination regime was associated with depletion of prostaglandin E2, a key downstream signaling molecule of Cox-2, caspase activation and downregulation of vascular endothelial growth factor in the tumor tissues. These data show that AITC and celecoxib complement each other for inhibition of bladder cancer and provide a novel combination approach for potential use for prevention or treatment of human bladder cancer.


Assuntos
Proliferação de Células/efeitos dos fármacos , Inibidores de Ciclo-Oxigenase 2/farmacologia , Isotiocianatos/farmacologia , Músculo Esquelético/efeitos dos fármacos , Pirazóis/farmacologia , Sulfonamidas/farmacologia , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Protocolos de Quimioterapia Combinada Antineoplásica , Western Blotting , Caspases/metabolismo , Celecoxib , Células Cultivadas , Dinoprostona/metabolismo , Endotélio Vascular/citologia , Endotélio Vascular/efeitos dos fármacos , Endotélio Vascular/metabolismo , Feminino , Conservantes de Alimentos/farmacologia , Humanos , Músculo Esquelético/metabolismo , Músculo Esquelético/patologia , Invasividade Neoplásica , Ratos , Ratos Endogâmicos F344 , Neoplasias da Bexiga Urinária/metabolismo , Neoplasias da Bexiga Urinária/patologia , Fator A de Crescimento do Endotélio Vascular/metabolismo
7.
Br J Nutr ; 109(1): 25-32, 2013 Jan 14.
Artigo em Inglês | MEDLINE | ID: mdl-22464629

RESUMO

Naturally occurring sulforaphane (SF) has been extensively studied for cancer prevention. However, little is known as to which organs may be most affected by this agent, which impedes its further development. In the present study, SF was administered to rats orally either in a single dose or once daily for 7 d. Tissue distribution of SF was measured by a HPLC-based method. Glutathione S-transferase (GST) and NAD(P)H:quinone oxidoreductase 1 (NQO1), two well-known cytoprotective phase 2 enzymes, were measured using biochemical assays to assess tissue response to SF. SF was delivered to different organs in vastly different concentrations. Tissue uptake of SF was the greatest in the stomach, declining rapidly in the descending gastro-intestinal tract. SF was rapidly eliminated through urinary excretion, and urinary concentrations of SF equivalents were 2-4 orders of magnitude higher than those of plasma. Indeed, tissue uptake level of SF in the bladder was second only to that in the stomach. Tissue levels of SF in the colon, prostate and several other organs were very low, compared to those in the bladder and stomach. Moreover, induction levels of GST and NQO1 varied by 3- to 6-fold among the organs of SF-treated rats, though not strictly correlated with tissue exposure to SF. Thus, there is profound organ specificity in tissue exposure and response to dietary SF, suggesting that the potential chemopreventive benefit of dietary SF may differ significantly among organs. These findings may provide a basis for prioritising organs for further chemopreventive study of SF.


Assuntos
Anticarcinógenos/metabolismo , Brassica/química , Mucosa Gástrica/metabolismo , Componentes Aéreos da Planta/química , Tiocianatos/metabolismo , Bexiga Urinária/metabolismo , Administração Oral , Animais , Anticarcinógenos/administração & dosagem , Anticarcinógenos/sangue , Anticarcinógenos/urina , Cromatografia Líquida de Alta Pressão , Indução Enzimática , Glutationa Transferase/biossíntese , Isotiocianatos , Cinética , Masculino , Desintoxicação Metabólica Fase II , NAD(P)H Desidrogenase (Quinona)/biossíntese , Especificidade de Órgãos , Distribuição Aleatória , Ratos , Ratos Endogâmicos F344 , Estômago/enzimologia , Sulfóxidos , Tiocianatos/administração & dosagem , Tiocianatos/sangue , Tiocianatos/urina , Distribuição Tecidual , Bexiga Urinária/enzimologia
8.
Carcinogenesis ; 33(2): 394-8, 2012 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-22131350

RESUMO

Naturally occurring allyl isothiocyanate (AITC) was recently shown to be selectively delivered to bladder cancer tissue via urinary excretion and to inhibit bladder cancer growth and muscle invasion in an animal model. AITC is excreted in urine mainly as N-acetyl-S-(N-allylthiocarbamoyl)cysteine, more commonly known as the N-acetylcysteine conjugate (NAC-AITC). We show here that treatment of human bladder cancer UM-UC-3 cells or rat bladder cancer AY-27 cells with NAC-AITC at 15 µM results in significant inhibition of cell growth and proliferation, together with cell cycle arrest and apoptosis. We also show that NAC-AITC administered orally at 10 µmol/kg body wt inhibits cancer growth by 40% and muscle invasion by 49% in an orthotopic rat bladder cancer model. Furthermore, the anticancer activity of NAC-AITC is associated with the modulation of several important molecular targets, including downregulation of both α-tubulin and ß-tubulin, activation of caspase-3 and downregulation of vascular endothelial growth factor. These results are similar to those shown previously for AITC and are consistent with the understanding that NAC-AITC is a carrier of AITC. Furthermore, comparison of the pharmacokinetic and physical properties of NAC-AITC with those of AITC suggests that NAC-AITC is superior to AITC for potential use for prevention and therapy of bladder cancer.


Assuntos
Cisteína/análogos & derivados , Isotiocianatos/farmacologia , Isotiocianatos/urina , Tiocarbamatos/farmacologia , Tiocarbamatos/urina , Neoplasias da Bexiga Urinária/tratamento farmacológico , Animais , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Cisteína/farmacologia , Cisteína/urina , Regulação para Baixo/efeitos dos fármacos , Feminino , Humanos , Invasividade Neoplásica , Ratos , Ratos Endogâmicos F344 , Tubulina (Proteína)/metabolismo , Neoplasias da Bexiga Urinária/patologia , Neoplasias da Bexiga Urinária/urina , Fator A de Crescimento do Endotélio Vascular/metabolismo
9.
J Exp Clin Cancer Res ; 41(1): 184, 2022 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-35650607

RESUMO

BACKGROUND: Epidermal growth factor receptor (EGFR) inhibitors, including cetuximab and panitumumab, are valuable therapeutics for colorectal cancer (CRC), but resistance to these inhibitors is common. The reason for such resistance is not well understood, which hampers development of better therapeutic strategies. Although activating mutations in KRAS, BRAF and PIK3CA are considered major drivers of CRC resistance to EGFR inhibitors, therapeutic targeting of these drug resistance drivers has not produced substantial clinical benefit. METHODS: We exploited cell lines and mouse tumor models (cell line xenografts and patient derived xenografts) for experiments of genetic and pharmacologic depletion of EGFR and/or its family member HER2, including EGFR mutants, inhibition of EGFR ligand shedding, and biochemical analysis of signaling proteins, to delineate the mechanism of CRC resistance to EGFR inhibitors and to assess the therapeutic activity of PEPDG278D, which is a recombinant human protein that induces the degradation of both EGFR and HER2. RESULTS: The sensitivity of CRC cells to cetuximab and panitumumab correlates with the ability of these drugs to induce EGFR downregulation. PEPDG278D strongly inhibits oncogenic signaling and growth of CRC cells by causing profound depletion of EGFR and HER2, regardless of activating mutations of KRAS, BRAF and PIK3CA. siRNA knockdown of EGFR or HER2 also inhibits CRC cells resistant to EGFR inhibitors. Tumors harboring mutated KRAS, BRAF and/or PIK3CA also overexpress EGFR ligands, further suggesting that EGFR signaling remains important to the tumors. While excessive tumor-generated high-affinity EGFR ligands block target engagement by PEPDG278D, aderbasib, an inhibitor of ADAM10 and ADAM17, enables PEPDG278D to exert strong antitumor activity by inhibiting ligand shedding. Moreover, adding fluorouracil, which is commonly used in CRC treatment, to the combination of PEPDG278D and aderbasib further enhances tumor inhibition. CONCLUSIONS: Our study shows that CRC resistance to EGFR inhibitors results primarily from the inability of the inhibitors to downregulate their target and that a PEPDG278D-based combination treatment overcomes the resistance.


Assuntos
Neoplasias Colorretais , Proteínas Proto-Oncogênicas B-raf , Animais , Cetuximab/farmacologia , Cetuximab/uso terapêutico , Classe I de Fosfatidilinositol 3-Quinases/metabolismo , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/genética , Neoplasias Colorretais/patologia , Resistencia a Medicamentos Antineoplásicos/genética , Receptores ErbB/metabolismo , Humanos , Ligantes , Camundongos , Panitumumabe/farmacologia , Panitumumabe/uso terapêutico , Inibidores de Proteínas Quinases/farmacologia , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/genética , Proteínas Proto-Oncogênicas B-raf/metabolismo , Proteínas Proto-Oncogênicas p21(ras)/metabolismo , Receptores Proteína Tirosina Quinases/metabolismo
10.
Commun Biol ; 4(1): 1373, 2021 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-34880421

RESUMO

Tumor suppressor p53, a critical regulator of cell fate, is frequently mutated in cancer. Mutation of p53 abolishes its tumor-suppressing functions or endows oncogenic functions. We recently found that p53 binds via its proline-rich domain to peptidase D (PEPD) and is activated when the binding is disrupted. The proline-rich domain in p53 is rarely mutated. Here, we show that oncogenic p53 mutants closely resemble p53 in PEPD binding but are transformed into tumor suppressors, rather than activated as oncoproteins, when their binding to PEPD is disrupted by PEPD knockdown. Once freed from PEPD, p53 mutants undergo multiple posttranslational modifications, especially lysine 373 acetylation, which cause them to refold and regain tumor suppressor activities that are typically displayed by p53. The reactivated p53 mutants strongly inhibit cancer cell growth in vitro and in vivo. Our study identifies a cellular mechanism for reactivation of the tumor suppressor functions of oncogenic p53 mutants.


Assuntos
Transformação Celular Neoplásica/genética , Dipeptidases/genética , Mutação , Proteína Supressora de Tumor p53/genética , Linhagem Celular , Dipeptidases/metabolismo , Humanos , Proteína Supressora de Tumor p53/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA