Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cancers (Basel) ; 14(3)2022 Jan 27.
Artigo em Inglês | MEDLINE | ID: mdl-35158903

RESUMO

Traditional oncological interventions have failed to improve survival for pancreatic cancer patients significantly. Novel treatment modalities able to release cancer-specific antigens, render immunologically "cold" pancreatic tumours "hot" and disrupt or reprogram the pancreatic tumour microenvironment are thus urgently needed. Therapeutic focused ultrasound exerts thermal and mechanical effects on tissue, killing cancer cells and inducing an anti-cancer immune response. The most important advances in therapeutic focused ultrasound use for initiation and augmentation of the cancer immunity cycle against pancreatic cancer are described. We provide a comprehensive review of the use of therapeutic focused ultrasound for the treatment of pancreatic cancer patients and describe recent studies that have shown an ultrasound-induced anti-cancer immune response in several tumour models. Published studies that have investigated the immunological effects of therapeutic focused ultrasound in pancreatic cancer are described. This article shows that therapeutic focused ultrasound has been deemed to be a safe technique for treating pancreatic cancer patients, providing pain relief and improving survival rates in pancreatic cancer patients. Promotion of an immune response in the clinic and sensitisation of tumours to the effects of immunotherapy in preclinical models of pancreatic cancer is shown, making it a promising candidate for use in the clinic.

2.
Int J Hyperthermia ; 38(1): 1443-1456, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34612127

RESUMO

BACKGROUND: Sub-ablative heat induces pleiotropic biological effects in cancer cells, activating programmed cell death or survival processes. These processes decide the fate of the heated cell. This study investigates these and assesses whether heat, in combination with HSP90 inhibition, augments cell death and induces a pro-immune phenotype in these cells. METHODS: HCT116 and HT29 cells were subjected to thermal doses (TID) of 60 and 120CEM43 using a PCR thermal cycler. HSP90 was inhibited with NVP-AUY922. Viability was assessed using the MTT assay. Cellular ATP and HSP70 release were assessed using ATP and Enzyme-linked Immunosorbent assays, respectively. Flow cytometry and immunoblotting were used to study the regulation of biomarkers associated with the heat shock response, the cell cycle, and immunogenic and programmed cell death. RESULTS: Exposure of HCT116 and HT29 cells to TIDs of 60 and 120CEM43 decreased their viability. In addition, treatment with 120CEM43 increased intracellular HSP70 and the percentage of HCT116/HT29 cells in the G2/M cell cycle phase, ATP release and Calreticulin/HSP70/HSP90 exposure in the plasma membrane, while downregulating CD47 compared to sham-exposed cells. When combined with NVP-AUY922, treatment of HCT116/HT29 cells with 120CEM43 resulted in a synergistic decrease of cell viability associated with the induction of apoptosis. Also, the combined treatments increased Calreticulin exposure, CD47 downregulation, and HSP70 release compared to the sham-exposed cells. CONCLUSION: Sub-ablative heating can act synergistically with the clinically relevant HSP90 inhibitor NVP-AUY922 to induce a pro-immunogenic form of cell death in colon cancer cells.


Assuntos
Neoplasias do Colo , Proteínas de Choque Térmico HSP90 , Apoptose , Linhagem Celular Tumoral , Neoplasias do Colo/tratamento farmacológico , Células HCT116 , Humanos
3.
J R Soc Interface ; 18(180): 20210266, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34229458

RESUMO

Pulsed high-intensity focused ultrasound (pHIFU) uses acoustic pressure to physically disrupt tumours. The aim of this study was to investigate whether pHIFU can be used in combination with immune checkpoint inhibitors (ICIs) to enhance survival of tumour-bearing animals. Murine orthotopic pancreatic KPC tumours were exposed both to a grid of pHIFU lesions (peak negative pressure = 17 MPa, frequency = 1.5 MHz, duty cycle = 1%, 1 pulse s-1, duration = 25 s) and to anti-CTLA-4/anti-PD-1 antibodies. Acoustic cavitation was detected using a weakly focused passive sensor. Tumour dimensions were measured with B-mode ultrasound before treatment and with callipers post-mortem. Immune cell subtypes were quantified with immunohistochemistry and flow cytometry. pHIFU treatment of pancreatic tumours resulted in detectable acoustic cavitation and increased infiltration of CD8+ T cells in the tumours of pHIFU and pHIFU + ICI-treated subjects compared with sham-exposed subjects. Survival of subjects treated with pHIFU + ICI was extended relative to both control untreated subjects and those treated with either pHIFU or ICI alone. Subjects treated with pHIFU + ICI had increased levels of CD8+IFNγ+ T cells, increased ratios of CD8+IFNγ+ to CD3+CD4+FoxP3+ and CD11b+Ly6G+ cells, and decreased CD11chigh cells in their tumours compared with controls. These results provide evidence that pHIFU combined with ICI may have potential for use in pancreatic cancer therapy.


Assuntos
Ablação por Ultrassom Focalizado de Alta Intensidade , Neoplasias Pancreáticas , Animais , Linfócitos T CD8-Positivos , Humanos , Inibidores de Checkpoint Imunológico , Camundongos , Pâncreas , Neoplasias Pancreáticas/tratamento farmacológico
4.
Int J Hyperthermia ; 36(1): 229-243, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-30700171

RESUMO

AIM: Thermal isoeffective dose (TID) has not been convincingly validated for application to predict biological effects from rapid thermal ablation (e.g., using >55 °C). This study compares the classical method of quantifying TID (derived from hyperthermia data) with a temperature-adjusted method based on the Arrhenius model for predicting cell survival in vitro, after either 'rapid' ablative or 'slow' hyperthermic exposures. METHODS: MTT assay viability data was obtained from two human colon cancer cell lines, (HCT116, HT29), subjected to a range of TIDs (120-720 CEM43) using a thermal cycler for hyperthermic (>2 minutes, <50 °C) treatments, or a novel pre-heated water bath based technique for ablative exposures (<10 seconds, >55 °C). TID was initially estimated using a constant RCEM>43°C=0.5, and subsequently using RCEM(T), derived from temperature dependent cell survival (injury rate) Arrhenius analysis. RESULTS: 'Slow' and 'rapid' exposures resulted in cell survival and significant regrowth (both cell lines) 10 days post-treatment for 240 CEM43 (RCEM>43°C=0.5), while 340-550 CEM43 (RCEM>43°C =0.5) delivered using 'rapid' exposures showed 12 ± 6% viability and 'slow' exposures resulted in undetectable viability. Arrhenius analysis of experimental data (activation energy ΔE = 5.78 ± 0.04 × 105 J mole-1, frequency factor A = 3.27 ± 11 × 1091 sec-1) yielded RCEM=0.42 * e0.0041*T which better-predicted cell survival than using R CEM> 43°C=0.5. CONCLUSIONS: TID calculated using an RCEM(T) informed by Arrhenius kinetic parameters provided a more consistent, heating strategy independent, predictor of cell viability, improving dosimetry of ablative thermal exposures. Cell viability was only undetectable above 305 ± 10 CEM43 using this revised measure.


Assuntos
Morte Celular/fisiologia , Calefação/métodos , Hipertermia Induzida/métodos , Humanos
5.
Int J Hyperthermia ; 31(5): 476-88, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25974074

RESUMO

PURPOSE: The pleiotropic effects of heat on cancer cells have been well documented. The biological effects seen depend on the temperature applied, and the heating duration. In this study we investigate the cytotoxic effects of heat on colon cancer cells and determine how different cell death processes such as autophagy, apoptosis and necroptosis play a role in cell response. MATERIALS AND METHODS: The thermal dose concept was used to provide a parameter that will allow comparison of different thermal treatments. Two human colon cancer cell lines, HCT116 and HT29, were subjected to ablative temperatures using a polymerase chain reaction thermal cycler. Temperature was recorded using thermocouples. Cell viability was assessed using the MTT assay. Induction of apoptosis was estimated using an enzyme-linked immunosorbent assay that detects cleaved cytoplasmic nucleosomes. Protein regulation was determined using immunoblotting. The percentage of cells undergoing apoptosis and autophagy was determined with annexin V/propidium iodide staining and a cationic amphiphilic tracer using fluorescence-activated cell sorting analysis. RESULTS: Exposure of colon cancer cells to ablative thermal doses results in decreased cell viability. The cytotoxic effect of heat is associated with induction of apoptosis and autophagy, the amount depending on both the thermal dose applied and on the time elapsed after treatment. Autophagy induction is mainly seen in live cells. RIPK3 protein levels are increased after exposure of cells to heat. A necroptosis inhibitor does not affect cell viability. CONCLUSIONS: Autophagy, apoptosis and necroptosis are associated with the response of these cancer cell lines to supra-normal temperatures.


Assuntos
Apoptose/genética , Autofagia/genética , Neoplasias do Colo/genética , Necrose/genética , Linhagem Celular Tumoral , Neoplasias do Colo/metabolismo , Neoplasias do Colo/mortalidade , Humanos , Taxa de Sobrevida
6.
Anticancer Agents Med Chem ; 14(8): 1169-78, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24954859

RESUMO

Lupulone, a ß-acid derived from hop extracts has been shown to exhibit cytotoxic activity against cancer cells. In this study we investigated the functional role of different modes of cell death that mediate anticancer effect of lupulone derivatives in prostate cancer cells. ELISA, immunoblotting and siRNA approaches were utilised to study cell death, expression of proteins of interest and their functional activities. We found that the anticancer effect of lupulone derivatives on prostate cancer cells is associated with induction of apoptosis and autophagy as determined by increases of DNA fragmentation and LC3I/ LC3II conversion respectively. Inhibition of apoptosis using a pan-caspase inhibitor resulted in increased levels of autophagy. Following screening of proteins associated with autophagy we found that Atg4ß expression was increased in prostate cancer cells after treatment with lupulone. Transfection of cells with siRNA against Atg4ß resulted in increased levels of apoptosis in prostate cancer cells. Treatment of prostate cancer cells with lupulone derivatives initiated two modes of cell death: apoptosis as a killing pathway and autophagy as a protection against cell death. Further studies are required to investigate the regulation of Atg4ß activity in lupulone derivatives-induced negative crosstalk between apoptosis and autophagy.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Neoplasias da Próstata/patologia , Terpenos/farmacologia , Clorometilcetonas de Aminoácidos/farmacologia , Antineoplásicos/química , Proteínas Relacionadas à Autofagia , Inibidores de Caspase/farmacologia , Caspases/metabolismo , Linhagem Celular Tumoral , Cisteína Endopeptidases/metabolismo , Fragmentação do DNA/efeitos dos fármacos , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Terpenos/química
7.
Nutr Cancer ; 65(7): 1086-92, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24168111

RESUMO

Lupulone, a ß-acid derived from hop extracts has been shown to exhibit antibacterial and anticancer activity. In this study we investigated the anticancer potency of lupulone and its novel derivatives and their mechanism of action on prostate cancer cells. Cell viability was determined using the MTT assay, and the ELISA approach was used to investigate induction of apoptosis. Immunoblot analysis was carried out to determine activation and regulation of proteins associated with cell death. Screening of natural and new lupulone derivatives for their anticancer activity demonstrated that one (lupulone derivative 1h) displayed stronger anticancer activity than lupulone itself on PC3 and DU145 prostate cancer cells. We further found that lupulone derivatives induced caspase-dependent apoptosis that is associated with activation of caspases 8, 9, and 3. Furthermore, caspase 8 inhibitor Z-IETD-fmk reduced cell death induced by lupulone derivatives, suggesting that apoptosis is mediated by caspase 8. Finally, we found that lupulone and its synthetic derivatives also increased formation of LC3II suggesting that autophagy is also implicated in prostate cancer cell death. The new lupulone derivatives induce caspase-dependent apoptosis and autophagy in prostate cancer cells and appear to be good candidates for further preclinical studies of prostate cancer treatment.


Assuntos
Antineoplásicos Fitogênicos/farmacologia , Humulus/química , Extratos Vegetais/farmacologia , Neoplasias da Próstata/metabolismo , Terpenos/farmacologia , Apoptose/efeitos dos fármacos , Autofagia/efeitos dos fármacos , Caspase 3/genética , Caspase 3/metabolismo , Caspase 8/genética , Caspase 8/metabolismo , Caspase 9/genética , Caspase 9/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Humanos , Masculino , Oligopeptídeos/farmacologia
8.
Methods Mol Biol ; 616: 149-59, 2010.
Artigo em Inglês | MEDLINE | ID: mdl-20379874

RESUMO

The endothelial cell plays a central role in the control of inflammatory processes. The recruitment of inflammatory leucocytes into the blood vessels is controlled by the expression of adhesion molecules on the endothelium as well as the secretion and presentation of chemokines. Indoleamine 2,3 dioxygenase (IDO) is an enzyme that metabolises tryptophan and is known to be central in the regulation of immune responses. IDO can be expressed on endothelial cells and can alter T-cell responses to the endothelium. We show that IDO can be readily transfected in primary endothelial cells and detail a method to determine the activity of the transfected protein.


Assuntos
Células Endoteliais/enzimologia , Indolamina-Pirrol 2,3,-Dioxigenase/metabolismo , Transfecção/métodos , Separação Celular , Células Cultivadas , Eletroporação , Células Endoteliais/citologia , Ensaios Enzimáticos , Humanos , Plasmídeos/genética , Veia Safena/citologia
9.
Pancreas ; 38(1): 78-84, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-19117086

RESUMO

OBJECTIVES: Pancreatic cancer is one of the leading causes of cancer related deaths in the western world. It is also resistant to most chemotherapeutic modalities. Phosphodiesterase-4 inhibitors (PDE4is) have found applications in the treatment of respiratory diseases. The aim of this study is to investigate the cytotoxic effect of 2 novel PDE4is, the CC-8075 and CC-8062 compounds in pancreatic cancer cells. METHODS: Cell proliferation was measured using the sulforhodamine B protein dye. Induction of apoptosis was detected using enzyme-linked immunosorbent assay. Regulation of proteins and posttranslational modifications were determined using immunoblotting. RESULTS: Treatment of pancreatic cancer cells with CC-8075 and CC-8062 reduces their proliferation and increases apoptosis that is caspase dependent in T3M4 cells. Furthermore, PDE4is increase phosphorylation of p38MAPK, mitogen-activated protein kinase (MAPK) kinase 3/6,MAPKYactivated protein kinase 2, Atf2, and Hsp27. The use of thep38MAPK-specific inhibitors SB202190 and SB203580 results in a modest reduction in PDE4i-induced apoptosis in T3M4 cells. Also, retinoids enhance apoptosis induced by CC-8075 and CC-8062 in GER cells. CONCLUSIONS: These results highlight the antiproliferative effects of the phosphodiesterase inhibitors CC-8075 and CC-8062 in pancreatic cancer cells and suggest that activation of p38MAPK signaling pathway may be associated with this process.


Assuntos
Antineoplásicos/farmacologia , Proliferação de Células/efeitos dos fármacos , Nucleotídeo Cíclico Fosfodiesterase do Tipo 4/farmacologia , Neoplasias Pancreáticas/patologia , Inibidores da Fosfodiesterase 4 , Inibidores de Fosfodiesterase/farmacologia , Alitretinoína , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , Humanos , Imidazóis/farmacologia , Neoplasias Pancreáticas/enzimologia , Fosforilação , Inibidores de Proteínas Quinases/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Tretinoína/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/antagonistas & inibidores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Int J Cancer ; 120(4): 743-52, 2007 Feb 15.
Artigo em Inglês | MEDLINE | ID: mdl-17131308

RESUMO

Doxycycline (DC) belongs to the tetracycline family of antibiotics and has been used clinically for over 5 decades. Despite advances in understanding the molecular pathogenesis of pancreatic cancer, no chemotherapy course has shown significant effectiveness. Hence new treatments are needed. In this study we report the pro-apoptotic effects of DC in 2 pancreatic adenocarcinoma cell lines, T3M4 and GER. Cell proliferation was measured using the SRB protein dye. Induction of apoptosis was detected using ELISA. Caspase activation was detected using either immunoblotting or a colorimetric assay based on cleavage of caspase-associated substrates. Expression of proteins and post-translational modifications were determined using immunoblotting. Treatment of pancreatic cancer cells with DC reduces their proliferation. This reduction is, at least partly, due to increased caspase-dependent apoptosis involving activation of caspase3, caspase7, caspase8, caspase9, caspase10 and increased levels of FADD. Inhibition of caspase8 or caspase10 but not caspase9 significantly decreases DC-induced apoptosis in both cell lines. Furthermore treatment of pancreatic cancer cells with DC increases protein levels of Bax and phosphorylation of members of the p38MAPK pathway such as p38MAPK, MKK3/6 and MAPKAPK2. These results provide an insight into mechanisms behind the pro-apoptotic effects of DC in pancreatic cancer cells.


Assuntos
Adenocarcinoma/patologia , Antibacterianos/farmacologia , Apoptose/efeitos dos fármacos , Caspases/metabolismo , Doxiciclina/farmacologia , Neoplasias Pancreáticas/patologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/metabolismo , Inibidores de Caspase , Proliferação de Células/efeitos dos fármacos , Ativação Enzimática/efeitos dos fármacos , Proteína de Domínio de Morte Associada a Fas/metabolismo , Humanos , Immunoblotting , Proteínas Quinases Ativadas por Mitógeno , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/metabolismo , Fosforilação/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Células Tumorais Cultivadas/efeitos dos fármacos
11.
Pancreas ; 32(1): 93-100, 2006 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-16340750

RESUMO

OBJECTIVES: Previous research has shown that the retinoid 9-cis retinoic acid (RA) promotes apoptosis in pancreatic cancer cells. The vitamin D analog EB1089 does not. Furthermore, cotreatment of cells with 9-cis RA and EB1089 abrogates apoptosis. To explain this, we studied the regulation of proteins involved in apoptotic signaling pathways in pancreatic cancer cells. METHODS: The pancreatic adenocarcinoma cell line T3M4 was used. Cell proliferation was measured using the SRB protein dye assay. Induction of apoptosis was evaluated using an ELISA assay. Caspase activation was detected using a colorimetric assay based on cleavage of a caspase-associated substrate. Regulation of protein levels and posttranslational events were detected using immunoblotting. RESULTS: We confirm that EB1089 diminishes apoptosis induced by 9-cis RA in T3M4 cells. We extend the study to show that EB1089 abrogates increases, induced by 9-cis RA, in caspase activation, p27Kip1 protein levels, Bim and Bax protein levels and in Bax/Bcl2 ratio. In addition, the CDKI p21Waf1 and CAII, a differentiation marker for pancreatic cancer cells are also differentially regulated. CONCLUSIONS: These results suggest that the inhibitory effects of EB1089 on 9-cis RA-induced apoptosis lie upstream of caspase activation and could be associated with reduction of p27Kip1 protein levels.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Calcitriol/análogos & derivados , Neoplasias Pancreáticas/tratamento farmacológico , Tretinoína/farmacologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Alitretinoína , Calcitriol/farmacologia , Caspases/metabolismo , Divisão Celular/efeitos dos fármacos , Linhagem Celular Tumoral , Inibidor de Quinase Dependente de Ciclina p27/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p27/metabolismo , Ensaio de Imunoadsorção Enzimática , Humanos , Neoplasias Pancreáticas/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...