Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 145
Filtrar
Mais filtros

Medicinas Complementares
Tipo de documento
Intervalo de ano de publicação
1.
Phytomedicine ; 48: 84-93, 2018 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-30195884

RESUMO

BACKGROUND: Glioblastomas (GBM) are one of the most aggressive tumor of the central nervous system with an average life expectancy of only 1-2 years after diagnosis, even with the use of advanced treatments with surgery, radiation, and chemotherapy. There are several anticancer drugs with alkylating properties that have been used in the therapy of malignant gliomas. Temozolomide (TMZ) is one of them, widely used even in combination with ionizing radiation. However, the main disadvantage of using these types of drugs in the treatment of GBM is the development of cancer drug resistance. Research of bioactive compounds with anticancer activity has been heavily explored. PURPOSE: This review focuses on a carotenoid and a phlorotannin present in seaweed, namely fucoxanthin and phloroglucinol, and their anticancer activity against glioblastoma. The combination of natural compounds with conventional drugs is also discussed. CONCLUSION: Several natural compounds existing in seaweeds, such as fucoxanthin and phoroglucinol, have shown cytotoxic activity in models in vitro and in vivo, acting through different molecular mechanisms, such as antioxidant, antiproliferative, DNA damage/DNA repair, proapoptotic, antiangiogenic and antimetastic. Within the scope of interactions with conventional drugs, there are evidences that some seaweed compounds could be used to potentiate the action of anticancer drugs. However, their effects and mechanisms of action, alone or in combination with anticancer drugs, namely TMZ, in glioblastoma cell, still few explored and require more attention due to the unquestionable high potential of these marine compounds.


Assuntos
Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Floroglucinol/farmacologia , Alga Marinha/química , Xantofilas/farmacologia , Antineoplásicos Alquilantes/farmacologia , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/patologia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Humanos , Temozolomida
2.
Cell Physiol Biochem ; 49(6): 2443-2462, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30261501

RESUMO

BACKGROUND/AIMS: Herbal materials derived from Juniperus communis (JCo) possess anticancer activity. In this study, we evaluated the efficacy of a JCo berry extract in suppressing glioblastoma growth. METHODS: The effects of JCo extract on the viability of normal and glioma cell lines was analyzed using a modified 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay. The synergistic therapeutic effect of JCo extract and temozolomide (TMZ) on glioma cells was examined by MTT analysis. Flow cytometry analysis, the terminal deoxynucleotidyl transferase-mediated nick end labeling (TUNEL) test, and western blotting were performed to identify the apoptotic pathway. To determine the in vivo efficacy of the JCo extract, rats were injected with 5 × 104 rat glioma RG2 cells in the back skin and brain hemisphere and then received a subcutaneous injection in the back skin that contained either JCo extract or vehicle. Finally, blood and histologic examinations were performed to evaluate JCo toxicity. RESULTS: The IC50 values of JCo extract were 57-69 µg/mL and 49-67 µg/mL in the glioblastoma cell lines after 24 and 48 h, respectively. However, in non-tumor cell lines, the respective IC50 values of JCo extract were 76-105 µg/mL and 77-108 µg/mL. The JCo extract had a stronger cytotoxicity and a larger range of IC50 values in glioma than in normal cells as compared to those effects caused by temozolomide (TMZ). In addition, the results of flow cytometry analysis, TUNEL test, and western blotting revealed that the JCo extract induced glioma cell cycle arrest through intrinsic and extrinsic apoptotic pathways. In the in vivo studies, a significant reduction of tumor size in JCo-treated rats, as measured by animal MRI, demonstrated that the JCo extract effectively inhibited glioma cell growth and successfully penetrated the blood-brain barrier. The immunohistochemical (IHC) staining detected positive signals of PCNA, VEGFR-1, and VEGFR -2 in 44.49%, 5.88%, and 5.85% of JCo-treated glioma cells, respectively. However, positive signals of PCNA, VEGFR-1, and VEGFR-2 were detected in 73.08%, 9.67%, and 11.70% of vehicle-treated glioma cells, respectively. The IHC examination of PCNA and VEGFR-1 and -2 indicated that JCo extract significantly decreased the degree of neovascularization. However, no significant differences in serum levels of blood cell count and hepatic enzymes, renal function index, and the histologic appearance of vital organs were detected between the JCo and vehicle-treated rats. CONCLUSION: The JCo extract penetrated the blood-brain barrier and significantly induced glioma cell apoptosis by reducing neovascularization via suppression of the PI3K/AKT/mTOR pathway. Furthermore, JCo extract was less cytotoxic to non-neoplastic vital organs than TMZ.


Assuntos
Apoptose/efeitos dos fármacos , Barreira Hematoencefálica/metabolismo , Juniperus/química , Extratos Vegetais/farmacologia , Animais , Barreira Hematoencefálica/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/patologia , Caspase 3/química , Caspase 3/metabolismo , Pontos de Checagem do Ciclo Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Sinergismo Farmacológico , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/patologia , Humanos , Juniperus/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Extratos Vegetais/química , Extratos Vegetais/uso terapêutico , Antígeno Nuclear de Célula em Proliferação/metabolismo , Temozolomida , Receptor 1 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo
3.
Phytomedicine ; 49: 41-51, 2018 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-30217261

RESUMO

BACKGROUND: Temozolomide (TMZ) is a first-line chemotherapeutic drug for malignant gliomas. Nonetheless, TMZ-induced side effects and drug resistance remain challenges. Our previous study showed the suppressive effects of honokiol on growth of gliomas. PURPOSE: This study was further aimed to evaluate if honokiol could enhance TMZ-induced insults toward malignant glioma cells and its possible mechanisms. METHODS: Human U87 MG glioma cells were exposed to TMZ, honokiol, and a combination of TMZ and honokiol. Cell survival, apoptosis, necrosis, and proliferation were successively assayed. Fluorometric substrate assays were conducted to determine activities of caspase-3, -6, -8, and -9. Levels of Fas ligand, Bax, and cytochrome c were immunodetected. Translocation of Bax to mitochondria were examined using confocal microscopy. Mitochondrial function was evaluated by assaying the mitochondrial membrane potential (MMP), reactive oxygen species (ROS), and complex I enzyme activity. Caspase-6 activity was suppressed using specific peptide inhibitors. The honokiol-induced effects were further confirmed using human U373 MG and murine GL261 cells. RESULTS: Exposure of human U87 MG glioma cells to honokiol significantly increased TMZ-induced DNA fragmentation and cell apoptosis. Interestingly, honokiol enhanced intrinsic caspase-9 activity without affecting extrinsic Fas ligand levels and caspase-8 activity. Sequentially, TMZ-induced changes in Bax translocation, the MMP, mitochondrial complex I enzyme activity, intracellular ROS levels, and cytochrome c release were enhanced by honokiol. Consequently, honokiol amplified TMZ-induced activation of caspases-3 and -6 in human U87 MG cells. Fascinatingly, suppressing caspase-6 activity concurrently decreased honokiol-induced DNA fragmentation and cell apoptosis. The honokiol-involved improvement in TMZ-induced intrinsic apoptosis was also confirmed in human U373 MG and murine GL261 glioma cells. CONCLUSIONS: This study showed that honokiol can enhance TMZ-induced apoptotic insults to glioma cells via an intrinsic mitochondrion-dependent mechanism. Our results suggest the therapeutic potential of honokiol to attenuate TMZ-induced side effects.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Dacarbazina/análogos & derivados , Medicamentos de Ervas Chinesas/farmacologia , Glioma/patologia , Lignanas/farmacologia , Mitocôndrias/fisiologia , Animais , Caspases/metabolismo , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Citocromos c/metabolismo , Fragmentação do DNA , Dacarbazina/farmacologia , Proteína Ligante Fas/metabolismo , Glioma/tratamento farmacológico , Humanos , Potencial da Membrana Mitocondrial , Camundongos , Espécies Reativas de Oxigênio/metabolismo , Transdução de Sinais/efeitos dos fármacos , Temozolomida
4.
Prog Neurol Surg ; 32: 180-195, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29990984

RESUMO

Alternating electric fields of intermediate frequencies, also known as Tumor Treating Fields (TTFields or TTF) is a novel anticancer treatment modality that disrupts tumor cell mitosis at the metaphase-anaphase transition, leading to mitotic catastrophe, aberrant mitotic exit, and/or cell death. It is realized through alteration of the cytokinetic cleavage furrow by interference of proteins possessing large dipole moments, like septin heterotrimer complex and α/ß-tubulin, and that results in disordered membrane contraction and failed cytokinesis. Aberrant mitotic exit also elicits immunogenic cell death, which may potentiate an immune response against treated tumors. Notably, in patients with recurrent glioblastoma multiforme (GBM) a prospective clinical trial demonstrated comparable overall survival and progression-free survival after TTFields therapy and best physician's choice chemotherapy. Moreover, it was shown that in patients with newly diagnosed GBM initially treated with standard chemoradiotherapy with daily temozolomide (TMZ), adjuvant TTFields combined with TMZ offered better survival than adjuvant TMZ alone. Therefore, TTFields therapy can be appreciated as a standard treatment option in cases of intracranial malignant gliomas, whereas future studies should establish its optimal combination with other existing anticancer modalities, which may offer additional survival benefits for patients.


Assuntos
Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/terapia , Terapia Combinada/métodos , Dacarbazina/análogos & derivados , Terapia por Estimulação Elétrica/métodos , Campos Eletromagnéticos , Glioma/terapia , Mitose , Recidiva Local de Neoplasia/terapia , Neoplasias Encefálicas/tratamento farmacológico , Dacarbazina/farmacologia , Glioma/tratamento farmacológico , Humanos , Recidiva Local de Neoplasia/tratamento farmacológico , Temozolomida
5.
Surg Pathol Clin ; 11(2): 297-311, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29751876

RESUMO

Drug-induced liver injury (DILI) is constantly changing as new drugs are approved and as new herbals and dietary supplements (HDS) reach the market. The pathologist plays a key role in the evaluation of DILI by classifying and interpreting the histologic findings considering patients' medical history and drug exposure. The liver biopsy findings may suggest alternative explanations of the injury and additional testing that should be performed to exclude non-DILI causes. Recent reports of iatrogenic liver injury are reviewed with attention to immunomodulatory and antineoplastic agents as well as reports of injury associated with HDS use.


Assuntos
Doença Hepática Induzida por Substâncias e Drogas/diagnóstico , Doença Hepática Induzida por Substâncias e Drogas/patologia , Antineoplásicos Alquilantes/efeitos adversos , Biópsia , Doença Hepática Induzida por Substâncias e Drogas/etiologia , Dacarbazina/efeitos adversos , Dacarbazina/análogos & derivados , Diagnóstico Diferencial , Suplementos Nutricionais/efeitos adversos , Hepatite Autoimune/diagnóstico , Hepatite Autoimune/patologia , Humanos , Imunossupressores/efeitos adversos , Fígado/patologia , Fitoterapia/efeitos adversos , Temozolomida
6.
Neurosci Bull ; 34(3): 573-588, 2018 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-29582250

RESUMO

In gliomas, the canonical Wingless/Int (WNT)/ß-catenin pathway is increased while peroxisome proliferator-activated receptor gamma (PPAR-γ) is downregulated. The two systems act in an opposite manner. This review focuses on the interplay between WNT/ß-catenin signaling and PPAR-γ and their metabolic implications as potential therapeutic target in gliomas. Activation of the WNT/ß-catenin pathway stimulates the transcription of genes involved in proliferation, invasion, nucleotide synthesis, tumor growth, and angiogenesis. Activation of PPAR-γ agonists inhibits various signaling pathways such as the JAK/STAT, WNT/ß-catenin, and PI3K/Akt pathways, which reduces tumor growth, cell proliferation, cell invasiveness, and angiogenesis. Nonsteroidal anti-inflammatory drugs, curcumin, antipsychotic drugs, adiponectin, and sulforaphane downregulate the WNT/ß-catenin pathway through the upregulation of PPAR-γ and thus appear to provide an interesting therapeutic approach for gliomas. Temozolomide (TMZ) is an antiangiogenic agent. The downstream action of this opposite interplay may explain the TMZ-resistance often reported in gliomas.


Assuntos
Neoplasias Encefálicas/metabolismo , Glioma/metabolismo , PPAR gama/metabolismo , Via de Sinalização Wnt/fisiologia , Animais , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Regulação para Baixo/efeitos dos fármacos , Glioma/terapia , Humanos , Temozolomida , Via de Sinalização Wnt/efeitos dos fármacos
7.
Mater Sci Eng C Mater Biol Appl ; 83: 44-50, 2018 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-29208287

RESUMO

Recently most of the researchers have turned their interest towards plant mediated synthesis of metal nanoparticles to avoid several environmental toxicants. In this manuscript, we have discussed the ecofriendly syntheses of zinc oxide nanoparticles (ZnO NPs) were achieved using Glycyrrhiza glabra (G. glabra) seed aqueous extract. The green synthesized ZnO NPs were characterized using analytical techniques like XRD, TEM, particle size histogram and Zeta potential. From the results, it was found that the green synthesized ZnO NPs were around 35nm in size with irregular spherical shape. The Zeta potential study of ZnO NPs was resulted to be high stabile with electronegative charge around -56.3mV. Further the G. glabra seed aqueous extract mediated synthesis of ZnO NPs were subjected to treat human glioblastoma cells with the help of temozolomide (TMZ) a commercially available drug by the method of MTT cell viability assay. The results stated that the ZnO NPs shows IC50 value around 30µg/mL results significantly. The plausible mechanism behind the mortality rate was also discussed in this manuscript.


Assuntos
Dacarbazina/análogos & derivados , Nanopartículas Metálicas/química , Óxido de Zinco/química , Linhagem Celular Tumoral , Dacarbazina/química , Dacarbazina/uso terapêutico , Glioma/tratamento farmacológico , Química Verde/métodos , Humanos , Extratos Vegetais/química , Folhas de Planta/química , Temozolomida , Óxido de Zinco/uso terapêutico
8.
JAMA ; 318(23): 2306-2316, 2017 12 19.
Artigo em Inglês | MEDLINE | ID: mdl-29260225

RESUMO

Importance: Tumor-treating fields (TTFields) is an antimitotic treatment modality that interferes with glioblastoma cell division and organelle assembly by delivering low-intensity alternating electric fields to the tumor. Objective: To investigate whether TTFields improves progression-free and overall survival of patients with glioblastoma, a fatal disease that commonly recurs at the initial tumor site or in the central nervous system. Design, Setting, and Participants: In this randomized, open-label trial, 695 patients with glioblastoma whose tumor was resected or biopsied and had completed concomitant radiochemotherapy (median time from diagnosis to randomization, 3.8 months) were enrolled at 83 centers (July 2009-2014) and followed up through December 2016. A preliminary report from this trial was published in 2015; this report describes the final analysis. Interventions: Patients were randomized 2:1 to TTFields plus maintenance temozolomide chemotherapy (n = 466) or temozolomide alone (n = 229). The TTFields, consisting of low-intensity, 200 kHz frequency, alternating electric fields, was delivered (≥ 18 hours/d) via 4 transducer arrays on the shaved scalp and connected to a portable device. Temozolomide was administered to both groups (150-200 mg/m2) for 5 days per 28-day cycle (6-12 cycles). Main Outcomes and Measures: Progression-free survival (tested at α = .046). The secondary end point was overall survival (tested hierarchically at α = .048). Analyses were performed for the intent-to-treat population. Adverse events were compared by group. Results: Of the 695 randomized patients (median age, 56 years; IQR, 48-63; 473 men [68%]), 637 (92%) completed the trial. Median progression-free survival from randomization was 6.7 months in the TTFields-temozolomide group and 4.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.52-0.76; P < .001). Median overall survival was 20.9 months in the TTFields-temozolomide group vs 16.0 months in the temozolomide-alone group (HR, 0.63; 95% CI, 0.53-0.76; P < .001). Systemic adverse event frequency was 48% in the TTFields-temozolomide group and 44% in the temozolomide-alone group. Mild to moderate skin toxicity underneath the transducer arrays occurred in 52% of patients who received TTFields-temozolomide vs no patients who received temozolomide alone. Conclusions and Relevance: In the final analysis of this randomized clinical trial of patients with glioblastoma who had received standard radiochemotherapy, the addition of TTFields to maintenance temozolomide chemotherapy vs maintenance temozolomide alone, resulted in statistically significant improvement in progression-free survival and overall survival. These results are consistent with the previous interim analysis. Trial Registration: clinicaltrials.gov Identifier: NCT00916409.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Dacarbazina/análogos & derivados , Terapia por Estimulação Elétrica , Glioblastoma/tratamento farmacológico , Adulto , Idoso , Antineoplásicos Alquilantes/efeitos adversos , Quimiorradioterapia , Dacarbazina/efeitos adversos , Dacarbazina/uso terapêutico , Intervalo Livre de Doença , Feminino , Seguimentos , Glioblastoma/radioterapia , Glioblastoma/cirurgia , Humanos , Quimioterapia de Manutenção , Masculino , Pessoa de Meia-Idade , Mitose , Análise de Sobrevida , Temozolomida
9.
BMJ Open ; 7(11): e017387, 2017 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-29102988

RESUMO

OBJECTIVE: To assess the efficacy and cost-effectiveness of modulated electrohyperthermia (mEHT) concurrent to dose-dense temozolomide (ddTMZ) 21/28 days regimen versus ddTMZ 21/28 days alone in patients with recurrent glioblastoma (GBM). DESIGN: A cohort of 54 patients with recurrent GBM treated with ddTMZ+mEHT in 2000-2005 was systematically retrospectively compared with five pooled ddTMZ 21/28 days cohorts (114 patients) enrolled in 2008-2013. RESULTS: The ddTMZ+mEHT cohort had a not significantly improved mean survival time (mST) versus the comparator (p=0.531) after a significantly less mean number of cycles (1.56 vs 3.98, p<0.001). Effect-to-treatment analysis (ETA) suggests that mEHT significantly enhances the efficacy of the ddTMZ 21/28 days regimen (p=0.011), with significantly less toxicity (no grade III-IV toxicity vs 45%-92%, p<0.0001). An estimated maximal attainable median survival time is 10.10 months (9.10-11.10). Cost-effectiveness analysis suggests that, unlike ddTMZ 21/28 days alone, ddTMZ+mEHT is cost-effective versus the applicable cost-effectiveness thresholds €US$25 000-50 000/quality-adjusted life year (QALY). Budget impact analysis suggests a significant saving of €8 577 947/$11 201 761 with 29.1-38.5 QALY gained per 1000 patients per year. Cost-benefit analysis suggests that mEHT is profitable and will generate revenues between €3 124 574 and $6 458 400, with a total economic effect (saving+revenues) of €5 700 034 to $8 237 432 per mEHT device over an 8-year period. CONCLUSIONS: Our ETA suggests that mEHT significantly improves survival of patients receiving the ddTMZ 21/28 days regimen. Economic evaluation suggests that ddTMZ+mEHT is cost-effective, budget-saving and profitable. After confirmation of the results, mEHT could be recommended for the treatment of recurrent GBM as a cost-effective enhancer of ddTMZ regimens, and, probably, of the regular 5/28 days regimen. mEHT is applicable also as a single treatment if chemotherapy is impossible, and as a salvage treatment after the failure of chemotherapy.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Glioblastoma/terapia , Hipertermia Induzida/economia , Adulto , Idoso , Antineoplásicos Alquilantes/economia , Neoplasias Encefálicas/economia , Análise Custo-Benefício , Dacarbazina/economia , Dacarbazina/uso terapêutico , Feminino , Alemanha , Glioblastoma/economia , Humanos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/terapia , Anos de Vida Ajustados por Qualidade de Vida , Análise de Regressão , Estudos Retrospectivos , Temozolomida
10.
Anticancer Res ; 37(11): 6025-6030, 2017 11.
Artigo em Inglês | MEDLINE | ID: mdl-29061781

RESUMO

BACKGROUND: Temozolomide is the primary chemotherapeutic agent used to treat glioblastoma. However, many tumors are initially resistant to or develop resistance to temozolomide, mainly due to high levels of O6-methylguanine DNA transferase (MGMT) which repairs DNA damage traditionally caused by temozolomide. Dihydrotanshinone (DHT) is extracted from Salvia miltiorrhiza, a Chinese medicinal plant, and has also been shown to have antiproliferative effects on various cancer cell lines. DHT has been to shown to induce apoptosis via induction endoplasmic reticulum stress, that can reportedly sensitize cells to temozolomide. MATERIALS AND METHODS: MTS cellular proliferation assays or trypan blue viability assays were used to determine the effects of DHT/temozolomide combinatorial treatment. Enzyme-linked immunosorbent assay (ELISA) was used to determine effects on MGMT and P-glycoprotein levels after singular and combinatorial treatment. RESULTS: DHT had a synergistic oncolytic effect in a MGMT-deficient cell line and a sensitizing effect in a MGMT-expressing cell line. Cytotoxicity due to DHT was shown to be reactive oxygen species-dependent, while the combinatorial effect of DHT and temozolomide synergistically reduced MGMT and P-glycoprotein levels. CONCLUSION: DHT was shown to augment temozolomide efficacy, indicating that, since DHT can penetrate the blood-brain barrier, temozolomide in combination with DHT may represent a promising therapeutic option for glioblastoma.


Assuntos
Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Sinergismo Farmacológico , Glioblastoma/patologia , Fenantrenos/farmacologia , Antineoplásicos Alquilantes/farmacologia , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Dacarbazina/farmacologia , Furanos , Glioblastoma/tratamento farmacológico , Humanos , Técnicas In Vitro , Quinonas , Temozolomida , Células Tumorais Cultivadas
11.
Mol Med Rep ; 16(6): 9401-9408, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-29039593

RESUMO

Malignant gliomas are a group of aggressive neoplasms among human cancers. The curative effects of current treatments are finite for improving the prognosis of patients. Hyperthermia (HT) is an effective treatment for cancers; however, the effects of HT with different temperatures in treatment of MG and relevant mechanisms remain unclear. MTT assay and Annexin V­fluorescein isothiocyanate/propidium iodide staining were used for investigating the proliferation and apoptosis of C6 cells, respectively. Western blotting was applied to detect the expression of proteins. Ultrasonography was employed to evaluate the tumor formation rate, growth rate, angiogenesis rate and degree of hardness of tumors in vivo. The authors certified that HT with 42­46˚C x 1 h, 1 t could inhibit proliferation, promote apoptosis, reduce tumor formation rate, growth rate, angiogenesis rate, degree of hardness of tumors, ischemic tolerance and anoxic tolerance, and have synergy with temozolomide in C6 cells. Long­term HT (43˚C x 1 h, 1 t/5 d, 90 d) did not cut down the sensitivity of C6 cells to HT, and sustainably inhibited the proliferation of C6 cells. Furthermore, the authors proved HT produced these effects primarily through inhibition of the EGFR/STAT3/HIF­1A/VEGF­A pathway.


Assuntos
Apoptose , Neoplasias Encefálicas/patologia , Receptores ErbB/metabolismo , Glioma/patologia , Hipertermia Induzida , Fator de Transcrição STAT3/metabolismo , Transdução de Sinais , Temperatura , Animais , Apoptose/efeitos dos fármacos , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/metabolismo , Carcinogênese/efeitos dos fármacos , Carcinogênese/patologia , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Dacarbazina/uso terapêutico , Glioma/tratamento farmacológico , Glioma/metabolismo , Células Endoteliais da Veia Umbilical Humana , Humanos , Ratos Wistar , Padrões de Referência , Transdução de Sinais/efeitos dos fármacos , Temozolomida
12.
J Neurooncol ; 135(3): 545-552, 2017 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-28849310

RESUMO

We characterized health-related quality of life (HRQoL), cognitive, and functional status in newly diagnosed glioblastoma (GBM) patients receiving Tumor treating fields (TTFields) with temozolomide (TMZ) versus TMZ alone in a planned interim analysis of a randomized phase III trial [NCT00916409], which showed significant improvement in progression-free and overall survival with TTFields/TMZ. After radiotherapy with concomitant TMZ, newly diagnosed GBM patients were randomized (2:1) to TTFields/TMZ (n = 210) or TMZ (n = 105). Interim analysis was performed in 315 patients with ≥18 months of follow-up. HRQoL, a secondary endpoint, was evaluated in per-protocol patient population and expressed as change from baseline (CFB) at 3, 6, and 9 months for each subscale in the EORTC QLQ-C30/BN20. Karnofsky performance scores (KPS) and Mini-Mental State Examination scores (MMSE) were assessed. CFB in HRQoL was balanced in treatment groups at the 12-month time point. Initially, HRQoL improved in patients treated with TTFields/TMZ (CFB3: 24% and CFB6: 13%) versus TMZ (CFB3: -7% and CFB6: -17%), though this difference was no longer evident at the 9-month point. General scales, including physical and social functioning, showed no difference at 9 and 12 months. TTFields/TMZ group reported higher concerns of "itchy skin". KPS over 12 months was just below 90 in both groups. Cognitive status (MMSE) was stable over time. HRQoL, KPS, and MMSE were balanced in both groups over time. There was no preliminary evidence that HRQoL, cognitive, and functional status is adversely affected by the continuous use of TTFields.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Terapia por Estimulação Elétrica , Glioblastoma/terapia , Qualidade de Vida , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/fisiopatologia , Neoplasias Encefálicas/psicologia , Cognição/efeitos dos fármacos , Terapia Combinada , Estudos Cross-Over , Dacarbazina/uso terapêutico , Terapia por Estimulação Elétrica/efeitos adversos , Terapia por Estimulação Elétrica/métodos , Feminino , Seguimentos , Glioblastoma/fisiopatologia , Glioblastoma/psicologia , Humanos , Avaliação de Estado de Karnofsky , Masculino , Testes de Estado Mental e Demência , Pessoa de Meia-Idade , Análise de Sobrevida , Temozolomida , Resultado do Tratamento , Adulto Jovem
13.
Chin Clin Oncol ; 6(4): 41, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28841803

RESUMO

BACKGROUND: Tumor treating fields (TTF, Optune®), one of the low-intensity alternating electric fields, have been demonstrated to disrupt mitosis and inhibit tumor growth with antimitotic properties in a variety of tumor types. The Food and Drug Administration (FDA) of the United States approved TTF for recurrent GBM and newly diagnosed GBM in 2011 and 2015, respectively. METHODS: A systematic review was conducted regarding the relevant studies published between January 1, 2000, and May 31, 2017 in PubMed database. The search term included "Tumor Treating Fields", "Optune", "TTF", "Novocure", and "GBM". This review summarizes the mechanism of action, efficacy, and adverse events based on pre-clinical studies and clinical trials for TTF in GBM. RESULTS: Pre-clinical studies showed that TTF could inhibit tumor growth in vitro and in vivo by disrupting mitosis, inducing cell cycle arrest and apoptosis. Two randomized phase III trials evaluated the efficacy and safety of TTF in GBM patients. It was revealed that the combination of TTF and standard chemotherapy (temozolomide) prolonged the progression-free survival (PFS) and overall survival (OS) without systemic safety issues in newly diagnosed GBM (EF-14 trial). For recurrent GBM, the efficacy of TTF monotherapy was shown to be equivalent in PFS and OS without systemic adverse events when compared to the control group that received best physicians-chosen chemotherapies (EF-11 trial). CONCLUSIONS: The advantages of TTF in GBM treatment, including non-invasive antitumor effect, superior therapeutic benefit in combination with chemotherapy, and minimal systematic toxicity, have been demonstrated in pre-clinical data and randomized phased III clinical trials. Future investigations will be needed to explore combinations of chemotherapy, radiation therapy, targeted therapy, as well as immunotherapy with this novel anti-tumor treatment modality to achieve additive or synergistic therapeutic benefit for GBM and other solid tumors.


Assuntos
Neoplasias Encefálicas/terapia , Terapia por Estimulação Elétrica/métodos , Glioblastoma/terapia , Antineoplásicos Alquilantes/uso terapêutico , Ensaios Clínicos Fase III como Assunto , Terapia Combinada/métodos , Dacarbazina/análogos & derivados , Dacarbazina/uso terapêutico , Intervalo Livre de Doença , Humanos , Ensaios Clínicos Controlados Aleatórios como Assunto , Segurança , Temozolomida , Resultado do Tratamento , Estados Unidos
14.
Nutr Cancer ; 69(6): 873-880, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28718668

RESUMO

Unmethylated O6-methylguanine-DNA-methyltransferase (MGMT) promoter leads to Temozolomide (TMZ) resistance in most of the glioblastoma multiforme (GBM) patients. We previously investigated the synergistic effect of Olea europaea leaf extract (OLE) on TMZ cytotoxicity through modulating microRNA expression. To date, knowledge about the effect of OLE on MGMT methylation is insufficient. The aim of the current study was to evaluate the potential modulating effect of OLE on the TMZ response of GBM tumors through MGMT methylation. Exposure to 1 mg/mL OLE caused a significant induction of CpG island methylation in the MGMT gene using Methyl quantitative PCR assay (P < 0.001). In WST-1 analysis, the use of 350 µM TMZ plus 1 mg/mL OLE significantly increased the TMZ response of MGMT unmethylated cells (P = 0.003). Using the comet assay, the impact of 1 mg/mL OLE plus 350 µM TMZ on the formation of DNA strand breaks was significantly higher than that of 450 µM TMZ alone (P < 0.001) and Western blot analysis revealed that, when cells are treated with 1-mg/mL OLE, the total p53 protein levels tended to decrease. The results presented in this study uniquely demonstrated that OLE synergistically increased the TMZ response of GBM tumors by regulating MGMT gene methylation and p53 expression. However, further studies to validate our findings are required.


Assuntos
Metilação de DNA , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Dacarbazina/análogos & derivados , Glioblastoma/tratamento farmacológico , Extratos Vegetais/farmacologia , Proteína Supressora de Tumor p53/metabolismo , Proteínas Supressoras de Tumor/metabolismo , Idoso , Linhagem Celular Tumoral , Ensaio Cometa , Ilhas de CpG , Dano ao DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Dacarbazina/uso terapêutico , Relação Dose-Resposta a Droga , Resistencia a Medicamentos Antineoplásicos/genética , Feminino , Humanos , Concentração Inibidora 50 , Masculino , Pessoa de Meia-Idade , Olea/química , Folhas de Planta/química , Regiões Promotoras Genéticas , Temozolomida , Proteína Supressora de Tumor p53/genética , Proteínas Supressoras de Tumor/genética
15.
Biomed Pharmacother ; 92: 254-264, 2017 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-28551545

RESUMO

Glioblastoma multiforme (GBM) is the most malignant brain cancer that causes high mortality in humans. It responds poorly to the most common cancer treatments, such as surgery, chemo- and radiation therapy. Temozolomide (TMZ) is an alkylating agent that has been widely used to treat GBM; resistance to this drug is often found. One unexplored possibility for overcoming this resistance is a treatment based on concomitant exposure to electromagnetic fields (EMF) and TMZ. Indeed, many evidences show that EMF affects cancer cells and drug performance. In this study, we evaluated the potential synergistic effect of 100µM TMZ and EMF (100Hz, 100G) on two human glioma cells line, i.e., U87 and T98G above single treatments, TMZ or EMF. Co-treatment synergistically enhanced apoptosis in U87 and T98G cells, by increasing the expression of P53, Bax, and Caspase-3 and decreasing that of Bcl-2 and Cyclin-D1. We also observed an increase in reactive oxygen species (ROS) production and the overexpression of the heme oxygenase-1 (HO-1) gene in comparison to controls. In conclusion, since EMF enhanced the apoptotic effect of TMZ, possibly through a redox regulation mechanism, the TMZ/EMF combination may be effective for glioma cancer treating. Further studies are needed to reveal the action mechanism of this possible novel therapeutic approach.


Assuntos
Antineoplásicos Alquilantes/toxicidade , Citotoxinas/toxicidade , Dacarbazina/análogos & derivados , Campos Eletromagnéticos/efeitos adversos , Glioblastoma/tratamento farmacológico , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/fisiologia , Dacarbazina/toxicidade , Glioblastoma/patologia , Humanos , Magnetoterapia/métodos , Temozolomida
16.
CNS Oncol ; 6(3): 185-193, 2017 07.
Artigo em Inglês | MEDLINE | ID: mdl-28399638

RESUMO

BACKGROUND: This post hoc analysis of the EF-14 trial (NCT00916409) of tumor-treating fields (TTFields) plus temozolomide versus temozolomide alone in newly diagnosed glioblastoma compared the efficacy of TTFields plus chemotherapy (physician's choice) versus chemotherapy alone after first recurrence. METHODS: Patients on TTFields plus temozolomide continued TTFields plus second-line chemotherapy after first recurrence. Some patients on temozolomide alone crossed over after approval of TTFields for recurrent GBM. The primary efficacy outcome was overall survival (OS). RESULTS: After disease progression, 131 patients received TTFields plus chemotherapy and 73 chemotherapy alone. Thirteen patients in the original temozolomide-alone group crossed over to receive TTFields plus chemotherapy after disease progression, resulting in 144 patients receiving TTFields plus chemotherapy and 60 chemotherapy alone. Median follow-up was 12.6 months. Bevacizumab, alone or with cytotoxic chemotherapy, was the most frequent treatment. Median OS in the TTFields plus chemotherapy group was significantly longer versus chemotherapy alone (11.8 vs 9.2 months; HR: 0.70; 95% CI, 0.48-1.00; p=0.049). TTFields showed a low toxicity safety profile, as previously reported, with no grade 3/4 device-related adverse events. CONCLUSION: TTFields plus chemotherapy after first disease recurrence on TTFields plus temozolomide or temozolomide alone prolonged OS in patients in the EF-14 trial.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Dacarbazina/análogos & derivados , Terapia por Estimulação Elétrica , Glioblastoma/terapia , Recidiva Local de Neoplasia/terapia , Adulto , Assistência ao Convalescente , Idoso , Idoso de 80 Anos ou mais , Antineoplásicos Imunológicos/uso terapêutico , Bevacizumab/uso terapêutico , Terapia Combinada , Dacarbazina/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Procedimentos Neurocirúrgicos , Radioterapia , Análise de Sobrevida , Temozolomida , Resultado do Tratamento , Adulto Jovem
18.
Phytother Res ; 31(5): 729-739, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28240396

RESUMO

Glioblastoma multiforme (GBM) is the most frequent, lethal and aggressive tumour of the central nervous system (CNS) in adults. Multidrug resistance (MDR) results in undesirable prognosis during GBM chemotherapy. In this study, we determined that Radicol (RAD), a novel trinorguaiane-type sesquiterpene originally isolated from the root of Dictamnus radicis Cortex, exhibited potently cytotoxic effect on temozolomide (TMZ)-resistant GBM cell lines in a dose-dependent manner. Radicol-induced apoptosis was confirmed with Hoechst 33342/propidium iodide and terminal deoxynucleotidyl transferase-mediated biotinylated UTP nick end-labelling (TUNEL) staining. Studies investigating the mechanism revealed that RAD triggered an attenuation of protein disulphide isomerase (PDI) and induced the unmitigated unfolded protein response (UPR) and lethal endoplasmic reticulum (ER) stress. Simultaneously, we further demonstrated that RAD suppressed the activation of Akt/mTOR/p70S6K phosphorylation by up-regulating the induction of glycogen synthase kinase-3ß (GSK-3ß). These results established a link between RAD-induced ER stress and inhibition of the Akt/mTOR/p70S6K pathway, and the attenuation of PDI and activation of GSK-3ß might be the synergistic target of antineoplastic effects during RAD-induced apoptosis. These findings suggested that RAD, possessing multiple cytotoxicity targets, low molecular weight and high lipid solubility, could be a promising agent for the treatment of malignant gliomas. Copyright © 2017 John Wiley & Sons, Ltd.


Assuntos
Antineoplásicos/farmacologia , Dacarbazina/análogos & derivados , Resistencia a Medicamentos Antineoplásicos , Glioma/patologia , Sesquiterpenos/farmacologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dacarbazina/farmacologia , Dictamnus/química , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Quinase 3 da Glicogênio Sintase/metabolismo , Humanos , Marcação In Situ das Extremidades Cortadas , Fosforilação/efeitos dos fármacos , Fitoterapia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Sesquiterpenos/química , Sesquiterpenos/classificação , Serina-Treonina Quinases TOR , Temozolomida
19.
J Nanosci Nanotechnol ; 17(2): 977-82, 2017 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-29671486

RESUMO

Solid lipid nanoparticles carrying a chemotherapeutic payload (i.e., temozolomide, TMZ) were synthesized using ghee, a clarified butter commonly used in traditional medicine and food products. Ghee solid lipid nanoparticles (GSLN) were characterized through dynamic light scattering, scanning electron microscopy, and UV-visible spectrometry. Formulations were generated with varying ratios of surfactant to lipid, resulting in a maximum TMZ entrapment efficiency of ˜70%. Optimal formulations were found to have an average size and polydispersity of ˜220 nm and 0.340, respectively. Release kinetics revealed TMZ-loaded GSLN (TMZ@GSLN) retained 10% of its pay-load at 2 h with ˜53% released in 5 h. Metabolic activity on human umbilical vein endothelial cells (HUVEC) revealed GSLN treatment resulted in an increase in viability following 3 d while treatment of glioblastoma LN-229 cells with TMZ@GSLN resulted in a significant decrease. Evaluation of diffusion of TMZ across a reconstructed HUVEC monolayer demonstrated TMZ@GSLN resulted in a significantly higher diffusion of drug when compared to free TMZ. This data suggests GSLN pose a promising delivery vehicle for TMZ-based therapeutics. Collectively, this data demonstrates GSLN exhibit favorable drug carrier properties with anti-proliferative properties in glioblastoma cancer cells.


Assuntos
Portadores de Fármacos , Ghee , Nanopartículas/química , Linhagem Celular Tumoral , Sobrevivência Celular/efeitos dos fármacos , Dacarbazina/análogos & derivados , Dacarbazina/química , Dacarbazina/farmacocinética , Dacarbazina/farmacologia , Portadores de Fármacos/química , Portadores de Fármacos/farmacocinética , Células Endoteliais da Veia Umbilical Humana , Humanos , Temozolomida
20.
Int J Nanomedicine ; 12: 29-40, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28031711

RESUMO

The anti-glioma effect of temozolomide (Tem) is sometimes undermined by the emerging resistance. Recently, resveratrol (Res), herbal medicine extracted from grape seeds, has been demonstrated for its potential use in chemosensitization. In the current study, both these drugs were loaded simultaneously into nanoparticles with methoxy poly(ethylene glycol)-poly epsilon caprolactone (mPEG-PCL) as drug carriers in order to achieve better antitumor efficiency. Tem/Res-coloaded mPEG-PCL nanoparticles were constructed, characterized, and tested for antitumor effect on glioma cells by using in vitro and xenograft model system. The nanoparticle constructs were satisfactory with drug loading content (Res =~12.4%; Tem =~9.3%) and encapsulation capacity of >85% for both the drugs. In addition, the coencapsulation led to better in vitro stability of the nanoparticles than Tem-loaded nanoparticles. An in vitro uptake study demonstrated a high uptake efficiency of the nanoparticles by glioma cells. The synergistic antitumor effect against glioma cells was observed in the combinational treatment of Res and Tem. Tem/Res-coloaded nanoparticles induced higher apoptosis in U87 glioma cells as compared to cells treated by the combination of free drugs. Tem/Res-coloaded particles caused more effective inhibition of phosphor-Akt, leading to upregulation of the downstream apoptotic proteins. In addition, the in vivo study showed the superior tumor delaying effect of coloaded nanoparticles than that of free drug combination. These results suggest that Tem/Res-coloaded nanoparticles could be a potential useful chemotherapeutic formulation for glioma therapy.


Assuntos
Antineoplásicos/farmacologia , Glioma/tratamento farmacológico , Animais , Antineoplásicos/administração & dosagem , Antineoplásicos Fitogênicos/administração & dosagem , Antineoplásicos Fitogênicos/farmacologia , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Dacarbazina/administração & dosagem , Dacarbazina/análogos & derivados , Dacarbazina/farmacologia , Portadores de Fármacos , Combinação de Medicamentos , Sinergismo Farmacológico , Glioma/metabolismo , Glioma/patologia , Humanos , Camundongos Nus , Nanopartículas , Tamanho da Partícula , Poliésteres , Polietilenoglicóis , Resveratrol , Estilbenos/administração & dosagem , Estilbenos/farmacologia , Temozolomida , Ensaios Antitumorais Modelo de Xenoenxerto
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA