Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 71
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Oncol Lett ; 26(5): 479, 2023 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-37818128

RESUMO

Ketogenic diets (KDs) are actively being evaluated for their potential anticancer effects. Although KDs are generally considered safe, their safety profile when combined with chemotherapy remains unknown. It is known that a KD enhances the anticancer effect of gemcitabine (2',2'-difluoro-2'-deoxycytidine) in LSL-KrasLSL-G12D/+Trp53R172H/+Pdx-1-Cre (KPC) tumor-bearing mice. In the present study, whether a KD in combination with gemcitabine affected the liver safety profile in KPC mice was evaluated. For this purpose, male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD; % kcal: 20% fat, 65% carbohydrate, 15% protein) + gemcitabine [control plus gemcitabine group (CG)] or a KD (% kcal: 84% fat, 15% protein, 1% carbohydrate) + gemcitabine [ketogenic plus gemcitabine group (KG)] for two months. After two months of treatment, no significant differences in body weight were observed between CGs and KGs. Moreover, the KD did not significantly alter the serum protein expression levels of liver enzymes, including aspartate aminotransferase, alanine aminotransferase and alkaline phosphatase. In addition, the KD did not alter markers of liver-lipid accumulation as well as serum cholesterol and triglyceride levels, compared with the CG-treated group. Upon histologic examination, steatosis was rare, with no notable differences between treatment groups. When examining liver fatty acid composition, KD treatment significantly increased the content of saturated fatty acids and significantly decreased levels of cis-monounsaturated fatty acids compared with the CG. Finally, the KD did not affect liver markers of inflammation and oxidative stress, nor the protein expression levels of enzymes involved in ketone bodies, such as 3-hydroxy-3-methylglutaryl-CoA lyase and hidroximetilglutaril-CoA sintasa, and glucose metabolism, such as hexokinase 2, pyruvate dehydrogenase and phosphofructokinase. In summary, a KD in combination with gemcitabine appears to be safe, with no apparent hepatotoxicity and these data support the further evaluation of a KD as an adjuvant dietary treatment for pancreatic cancer.

2.
Int J Mol Sci ; 24(13)2023 Jun 28.
Artigo em Inglês | MEDLINE | ID: mdl-37445930

RESUMO

Cancer-associated cachexia (CAC) is a critical contributor to pancreatic ductal adenocarcinoma (PDAC) mortality. Thus, there is an urgent need for new strategies to mitigate PDAC-associated cachexia; and the exploration of dietary interventions is a critical component. We previously observed that a ketogenic diet (KD) combined with gemcitabine enhances overall survival in the autochthonous LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx1-Cre (KPC) mouse model. In this study, we investigated the effect and cellular mechanisms of a KD in combination with gemcitabine on the maintenance of skeletal muscle mass in KPC mice. For this purpose, male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD), a KD, a CD + gemcitabine (CG), or a KD + gemcitabine (KG) group. We observed that a KD or a KG-mitigated muscle strength declined over time and presented higher gastrocnemius weights compared CD-fed mice. Mechanistically, we observed sex-dependent effects of KG treatment, including the inhibition of autophagy, and increased phosphorylation levels of eIF2α in KG-treated KPC mice when compared to CG-treated mice. Our data suggest that a KG results in preservation of skeletal muscle mass. Additional research is warranted to explore whether this diet-treatment combination can be clinically effective in combating CAC in PDAC patients.


Assuntos
Carcinoma Ductal Pancreático , Dieta Cetogênica , Neoplasias Pancreáticas , Camundongos , Masculino , Feminino , Animais , Gencitabina , Caquexia/tratamento farmacológico , Caquexia/etiologia , Neoplasias Pancreáticas/complicações , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Carcinoma Ductal Pancreático/patologia
3.
Food Funct ; 14(6): 2684-2697, 2023 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-36752162

RESUMO

Chlorogenic acid (CGA) and epigallocatechin-3-gallate (EGCG) are major polyphenolic constituents of coffee and green tea with beneficial health properties. In this study, we evaluated the gut protecting effect of CGA and EGCG, alone or in combination, on D-galactose-induced aging mice. CGA plus EGCG more effectively improved the cognition deficits and protected the gut barrier function, compared with the agents alone. Specifically, CGA plus EGCG prevented the D-galactose mediated reactive oxygen species accumulation by increasing the total antioxidant capacity, reducing the levels of malondialdehyde, and suppressing the activity of the antioxidant enzymes superoxide dismutase and catalase. In addition, supplementation of CGA and EGCG suppressed gut inflammation by reducing the levels of the proinflammatory cytokines TNFα, IFNγ, IL-1ß and IL-6. Moreover, CGA and EGCG modulated the gut microbiome altered by D-galactose. For instance, CGA plus EGCG restored the Firmicutes/Bacteroidetes ratio of the aging mice to control levels. Furthermore, CGA plus EGCG decreased the abundance of Lactobacillaceae, Erysipelotrichaceae, and Deferribacteraceae, while increased the abundance of Lachnospiraceae, Muribaculaceae, and Rikenellaceae, at the family level. In conclusion, CGA in combination with EGCG ameliorated the gut alterations induced by aging, in part, through antioxidant and anti-inflammatory effects, along with its gut microbiota modulatory capacity.


Assuntos
Antioxidantes , Catequina , Camundongos , Animais , Antioxidantes/farmacologia , Ácido Clorogênico/farmacologia , Galactose/efeitos adversos , Envelhecimento , Catequina/farmacologia
4.
Cancer Res Commun ; 2(9): 951-965, 2022 09.
Artigo em Inglês | MEDLINE | ID: mdl-36382086

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) continues to be a major health problem. A ketogenic diet (KD), characterized by a very low carbohydrate and high fat composition, has gained attention for its anti-tumor potential. We evaluated the effect and mechanisms of feeding a strict KD alone or in combination with gemcitabine in the autochthonous LSL-KrasG12D/+; LSL-Trp53 R172H/+; Pdx1-Cre (KPC) mouse model. For this purpose, both male and female pancreatic tumor-bearing KPC mice were allocated to a control diet (CD; %kcal: 70% carb, 14% protein, 16% fat), a KD (%kcal: 14% protein, 1% carb, 85% fat), a CD + gemcitabine (CG), or a KD + gemcitabine (KG) group. Mice fed a KD alone or in combination with gemcitabine showed significantly increased blood ß-hydroxybutyrate levels compared to mice fed a CD or CG. KPC mice fed a KG had a significant increase in overall median survival compared to KPC mice fed a CD (increased overall median survival by 42%). Interestingly, when the data was disaggregated by sex, the effect of a KG was significant in female KPC mice (60% increase in median overall survival), but not in male KPC mice (28% increase in median overall survival). Mechanistically, the enhanced survival response to a KD combined with gemcitabine was multifactorial, including inhibition of ERK and AKT pathways, regulation of fatty acid metabolism and the modulation of the gut microbiota. In summary, a KD in combination with gemcitabine appears beneficial as a treatment strategy in PDAC in KPC mice, deserving further clinical evaluation.


Assuntos
Carcinoma Ductal Pancreático , Dieta Cetogênica , Neoplasias Pancreáticas , Camundongos , Masculino , Feminino , Animais , Gencitabina , Neoplasias Pancreáticas/tratamento farmacológico , Carcinoma Ductal Pancreático/tratamento farmacológico , Neoplasias Pancreáticas
5.
Neoplasia ; 24(2): 133-144, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34968866

RESUMO

Novel therapeutic strategies are needed in the fight against pancreatic cancer. We have previously documented the chemopreventive effect of MDC-22 in preclinical models of pancreatic cancer. In the present work, we examined the therapeutic effects of MDC-22 in patient-derived tumor xenografts (PDTXs) and in LSL-KrasG12D/+, LSL-Trp53R172H/+, Pdx1-Cre (KPC) genetically engineered mice, two complementary and clinically relevant animal models of pancreatic cancer. In addition, we evaluated whether MDC-22 could synergize with current chemotherapeutic drugs used in the clinic. MDC-22 reduced the growth of various human pancreatic cancer cell lines in a concentration-dependent manner. In vivo, MDC-22 strongly reduced patient-derived pancreatic tumor xenograft growth by 50%, and extended survival of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mice by over a month (5.3 months versus 7.0 months). In both models, MDC-22 inhibited EGFR activation and its downstream signals, including ERK and FAK phosphorylation. In human pancreatic cancer cell lines, MDC-22 enhanced the growth inhibitory effect of irinotecan, and to a lesser degree those of gemcitabine and nab-paclitaxel. Normal human pancreatic epithelial cells were more resistant to the cytotoxic effects of, both, MDC-22 alone or in combination with irinotecan, indicating selectivity. Furthermore, MDC-22 enhanced irinotecan's effect on cell migration, in part, by inhibiting EGFR/FAK signaling. Collectively, our results indicate that MDC-22 is an effective anticancer drug in preclinical models of pancreatic cancer, and suggest that MDC-22 plus irinotecan as drug combination strategy for pancreatic cancer treatment, which warrants further evaluation.


Assuntos
Antineoplásicos/farmacologia , Aspirina/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Animais , Aspirina/análogos & derivados , Modelos Animais de Doenças , Quimioterapia Combinada , Receptores ErbB/antagonistas & inibidores , Humanos , Irinotecano/farmacologia , Camundongos , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , Resultado do Tratamento
6.
Cancer Res Commun ; 2(12): 1668, 2022 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-36970724

RESUMO

[This corrects the article DOI: 10.1158/2767-9764.CRC-22-0256.][This corrects the article DOI: 10.1158/2767-9764.CRC-22-0256.].

7.
Food Funct ; 12(22): 11671-11685, 2021 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-34730149

RESUMO

Cyclophosphamide (CTX) is an antitumor drug commonly used to treat various cancer types. Unfortunately, its toxic side effects, including gastrointestinal (GI) toxicity, affect treatment compliance and patients' prognosis. Thus, there is a critical need of evaluating strategies that may improve the associated GI toxicity induced by CTX. In this work, we evaluated the capacity of epigallocatechin-3-gallate (EGCG), a major constituent of green tea, to improve the recovery of gut injury induced by CTX in mice. Treatment with CTX for 5 days severely damaged the intestinal structure, increased immune-related cytokines (TNFα, IL-10 and IL-21), reduced the expression levels of tight junction proteins (ZO-1, occludin, claudin-1), induced reactive oxygen species, altered the composition of gut microbiota, and reduced short chain fatty acid levels. EGCG treatment, starting one day after the last CTX dose, significantly improved the intestinal structure, ameliorated gut permeability, and restored ZO-1, occludin and claudin-1 levels. Moreover, EGCG reduced TNFα, IL-10 and IL-21 levels and decreased oxidative stress by regulating the activities of the antioxidant enzymes catalase, superoxide dismutase and glutathione peroxidase. Finally, EGCG treatment restored the composition of gut microbiota and the levels of the short chain fatty acids. In conclusion, these findings indicate that EGCG may function as an effective bioactive compound to minimize CTX-induced GI tract toxicity.


Assuntos
Catequina/análogos & derivados , Ciclofosfamida/efeitos adversos , Microbioma Gastrointestinal/efeitos dos fármacos , Enteropatias/metabolismo , Junções Íntimas/efeitos dos fármacos , Animais , Catequina/farmacologia , Disbiose/metabolismo , Inflamação/metabolismo , Enteropatias/induzido quimicamente , Masculino , Camundongos , Camundongos Endogâmicos ICR
8.
Nutrients ; 13(9)2021 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-34579079

RESUMO

Pancreatic ductal adenocarcinoma (PDAC) is an aggressive and extremely therapy-resistant cancer. It is estimated that up to 80% of PDAC patients present with cachexia, a multifactorial disorder characterized by the involuntary and ongoing wasting of skeletal muscle that affects therapeutic response and survival. During the last decade, there has been an increased interest in exploring dietary interventions to complement the treatment of PDAC and associated cachexia. Ketogenic diets (KDs) have gained attention for their anti-tumor potential. Characterized by a very low carbohydrate, moderate protein, and high fat composition, this diet mimics the metabolic changes that occur in fasting. Numerous studies report that a KD reduces tumor growth and can act as an adjuvant therapy in various cancers, including pancreatic cancer. However, research on the effect and mechanisms of action of KDs on PDAC-associated cachexia is limited. In this narrative review, we summarize the evidence of the impact of KDs in PDAC treatment and cachexia mitigation. Furthermore, we discuss key cellular mechanisms that explain KDs' potential anti-tumor and anti-cachexia effects, focusing primarily on reprogramming of cell metabolism, epigenome, and the gut microbiome. Finally, we provide a perspective on future research needed to advance KDs into clinical use.


Assuntos
Adenocarcinoma/complicações , Adenocarcinoma/dietoterapia , Caquexia/etiologia , Dieta Cetogênica , Neoplasias Pancreáticas/complicações , Neoplasias Pancreáticas/dietoterapia , Humanos
9.
Nanomedicine (Lond) ; 16(25): 2291-2303, 2021 10.
Artigo em Inglês | MEDLINE | ID: mdl-34579548

RESUMO

Aim: To investigate a novel strategy to target tumor-associated macrophages and reprogram them to an antitumor phenotype in pancreatic adenocarcinoma (PDAC). Methods: M2 peptides were conjugated to HA-PEG/HA-PEI polymer to form self-assembled nanoparticles with miR-125b. The efficacy of HA-PEI/PEG-M2peptide nanoparticles in pancreatic tumors from LSL-KrasG12D/+, LSL-Trp53R172H/+, Pdx1-Cre genetically engineered mice was evaluated. Results:In vitro M2 macrophage-specific delivery of targeted nanoformulations was demonstrated. Intraperitoneal administration of M2-targeted nanoparticles showed preferential accumulation in the pancreas of KPC-PDAC mice and an above fourfold increase in the M1-to-M2 macrophage ratio compared with transfection with scrambled miR. Conclusion: M2-targeted HA-PEI/PEG nanoparticles with miR-125b can transfect tumor-associated macrophages in pancreatic tissues and may have implications for PDAC immunotherapy.


Assuntos
Adenocarcinoma , MicroRNAs , Nanopartículas , Neoplasias Pancreáticas , Macrófagos Associados a Tumor , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Animais , Ácido Hialurônico , Camundongos , MicroRNAs/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/genética , Transfecção
10.
Oncol Rep ; 45(4)2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33649787

RESUMO

Lung cancer (LC) and pancreatic cancer (PC) are the first and fourth leading causes of cancer­related deaths in the US. Deregulated cell cycle progression is the cornerstone for rapid cell proliferation, tumor development, and progression. Here, we provide evidence that a novel combinatorial miR treatment inhibits cell cycle progression at two phase transitions, through their activity on the CDK4 and CDK1 genes. Following transfection with miR­143 and miR­506, we analyzed the differential gene expression of CDK4 and CDK1, using qPCR or western blot analysis, and evaluated cell cycle inhibition, apoptosis and cytotoxicity. The combinatorial miR­143/506 treatment downregulated CDK4 and CDK1 levels, and induced apoptosis in LC cells, while sparing normal lung fibroblasts. Moreover, the combinatorial miR treatment demonstrated a comparable activity to clinically tested cell cycle inhibitors in inhibiting cell cycle progression, by presenting substantial inhibition at the G1/S and G2/M cell cycle transitions. More importantly, the miR­143/506 treatment presented a broader application, effectively downregulating CDK1 and CDK4 levels, and reducing cell growth in PC cells. These findings suggest that the miR­143/506 combination acts as a promising approach to inhibit cell cycle progression for cancer treatment with minimal toxicity to normal cells.


Assuntos
Quinases Ciclina-Dependentes/metabolismo , Neoplasias Pulmonares/metabolismo , MicroRNAs/metabolismo , Neoplasias Pancreáticas/metabolismo , Aminopiridinas/farmacologia , Antineoplásicos/farmacologia , Proteína Quinase CDC2/antagonistas & inibidores , Proteína Quinase CDC2/genética , Proteína Quinase CDC2/metabolismo , Ciclo Celular/efeitos dos fármacos , Ciclo Celular/fisiologia , Processos de Crescimento Celular , Linhagem Celular Tumoral , Quinase 4 Dependente de Ciclina/antagonistas & inibidores , Quinase 4 Dependente de Ciclina/genética , Quinase 4 Dependente de Ciclina/metabolismo , Quinase 6 Dependente de Ciclina/antagonistas & inibidores , Quinase 6 Dependente de Ciclina/genética , Quinase 6 Dependente de Ciclina/metabolismo , Quinases Ciclina-Dependentes/antagonistas & inibidores , Regulação para Baixo , Fibroblastos/citologia , Fibroblastos/metabolismo , Flavonoides/farmacologia , Humanos , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/terapia , MicroRNAs/administração & dosagem , MicroRNAs/genética , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Neoplasias Pancreáticas/terapia , Piperidinas/farmacologia , Purinas/farmacologia , Transfecção , Regulação para Cima
11.
Nutr Res ; 88: 1-18, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33607535

RESUMO

A significant number of pancreatic cancer cases are due to modifiable risk factors, with many being attributed to increased body fatness. This has sparked investigators to examine the role played by high dietary fat intake in pancreatic cancer development and the mechanisms driving this connection. However, there is currently no consensus on how dietary fat quantity and composition specifically affect pancreatic carcinogenesis. The objective of this narrative review is to discuss the link between high total fat consumption and fatty acid composition (saturated, mono-, or poly-unsaturated fats) with pancreatic cancer incidence and progression. Following our detailed analysis of the strengths and weaknesses of recent preclinical and human studies, we discuss existing research gaps and opportunities, and provide recommendations for future studies. Numerous studies suggest that diets high in omega-3 polyunsaturated fatty acids are associated with reduced pancreatic cancer risk. However, the current evidence appears insufficient for a general conclusion regarding the impact of other types of fat in pancreatic carcinogenesis, with many studies providing inconclusive findings due to study limitations. Thus, we recommend future studies to include detailed methodology of the animal experiments, not limited to the diet composition, type of ingredients, formulations, and administration of the diets. Moreover, human studies should include a diverse population and well-characterized biomarkers for accurate determination of dietary fat intake. Ultimately, this will aid the study rigor, and improve our understanding of the impact of fat quantity and composition in pancreatic carcinogenesis.


Assuntos
Gorduras na Dieta/efeitos adversos , Neoplasias Pancreáticas/epidemiologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Animais , Cricetinae , Gorduras na Dieta/administração & dosagem , Gorduras na Dieta/análise , Progressão da Doença , Ácidos Graxos/análise , Ácidos Graxos Ômega-3/administração & dosagem , Feminino , Humanos , Masculino , Camundongos , Pessoa de Meia-Idade , Neoplasias Pancreáticas/fisiopatologia , Fatores de Risco
12.
Arch Biochem Biophys ; 692: 108546, 2020 10 15.
Artigo em Inglês | MEDLINE | ID: mdl-32818507

RESUMO

Epigallocatechin-3-gallate (EGCG), a major polyphenol component of green tea, presents anticancer efficacy. However, its exact mechanism of action is not known. In this study, we evaluated the effect of EGCG alone or in combination with current chemotherapeutics [gemcitabine, 5-flourouracil (5-FU), and doxorubicin] on pancreatic, colon, and lung cancer cell growth, as well as the mechanisms involved in the combined action. EGCG reduced pancreatic, colon, and lung cancer cell growth in a concentration and time-dependent manner. EGCG strongly induced apoptosis and blocked cell cycle progression. Moreover, EGCG enhanced the growth inhibitory effect of 5-FU and doxorubicin. Of note, EGCG enhanced 5-FU's and doxorubicin's effect on apoptosis, but not on cell cycle. Mechanistically, EGCG reduced ERK phosphorylation concentration-dependently, and sensitized gemcitabine, 5-FU, and doxorubicin to further suppress ERK phosphorylation in multiple cancer cell lines. In conclusion, EGCG presents a strong anticancer effect in pancreatic, colon, and lung cancer cells and is a robust combination partner for multiple chemotherapeutics as evidenced by reducing cancer cell growth, in part, by inhibiting the ERK pathway.


Assuntos
Antineoplásicos/farmacologia , Apoptose/efeitos dos fármacos , Catequina/análogos & derivados , Citotoxinas/farmacologia , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Neoplasias/tratamento farmacológico , Células A549 , Catequina/farmacologia , Células HT29 , Humanos , Neoplasias/metabolismo , Neoplasias/patologia
13.
Biochem Pharmacol ; 175: 113923, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32217102

RESUMO

Colorectal cancer (CRC) is one of the most common cancers worldwide. Epidemiological studies indicate that consumption of fruits and vegetables containing procyanidins is associated with lower CRC risk. This study investigated the capacity of two dimeric procyanidins composed of epicatechin gallate (ECG) or epigallocatechin gallate (EGCG) isolated from persimmons, to inhibit CRC cell growth and promote apoptosis, characterizing the underlying mechanisms. ECG and EGCG dimers reduced the growth of five human CRC cell lines in a concentration (10-60 µM)- and time (24-72 h)-dependent manner, with a 72 h-IC50 value in Caco-2 cells of 10 and 30 µM, respectively. ECG and EGCG dimers inhibited Caco-2 cell proliferation by arresting the cell cycle in G2/M phase and by inducing apoptosis via the mitochondrial pathway. In addition, ECG and EGCG dimers inhibited cell migration, invasion, and adhesion, decreasing the activity of matrix metalloproteinases (MMP-2/9). Mechanistically, ECG and EGCG dimers inhibited the activation of lipid raft-associated epidermal growth factor (EGF) receptor (EGFR), without affecting its localization at lipid rafts. In particular, ECG and EGCG dimers reduced EGFR phosphorylation at Tyr1068 residue, prevented EGFR dimerization and activation upon stimulation, and induced EGFR internalization both in the absence and presence of EGF. Furthermore, ECG and EGCG dimers increased EGFR phosphorylation at Tyr1045 residue, providing a docking site for ubiquitin ligase c-Cbl and induced EGFR degradation by the proteasome. Downstream of EGFR, ECG and EGCG dimers inhibited the activation of the MEK/ERK1/2 and PI3K/AKT signaling pathways, downregulating proteins involved in the modulation of cell survival. In conclusion, ECG and EGCG dimers reduced CRC cell growth by inhibiting EGFR activation at multiple steps, including the disruption of lipid rafts integrity and promoting EGFR degradation. These results shed light on a potential molecular mechanism on how procyanidins-rich diets may lower CRC risk.


Assuntos
Catequina/análogos & derivados , Neoplasias Colorretais/metabolismo , Inibidores do Crescimento/farmacologia , Microdomínios da Membrana/metabolismo , Proantocianidinas/farmacologia , Anticarcinógenos/farmacologia , Anticarcinógenos/uso terapêutico , Antineoplásicos Fitogênicos/farmacologia , Antineoplásicos Fitogênicos/uso terapêutico , Células CACO-2 , Catequina/farmacologia , Catequina/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Relação Dose-Resposta a Droga , Receptores ErbB/antagonistas & inibidores , Receptores ErbB/metabolismo , Inibidores do Crescimento/uso terapêutico , Células HCT116 , Células HT29 , Humanos , Microdomínios da Membrana/efeitos dos fármacos , Proantocianidinas/uso terapêutico , Multimerização Proteica/efeitos dos fármacos , Multimerização Proteica/fisiologia , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia
14.
Carcinogenesis ; 41(7): 927-939, 2020 07 14.
Artigo em Inglês | MEDLINE | ID: mdl-31584613

RESUMO

New chemotherapeutic agents are needed for pancreatic cancer (PC). We have previously shown that phospho-valproic acid (MDC-1112) is effective in cell-line xenografts of PC. Here, we explored whether MDC-1112 is effective in additional clinically relevant animal models of PC and whether MDC-1112 enhances the anticancer effect of clinically used chemotherapeutic agents. MDC-1112 alone strongly reduced patient-derived pancreatic tumor xenograft growth, and extended survival of LSL-KrasG12D/+; LSL-Trp53R172H/+; Pdx1-Cre (KPC) mice. In both models, MDC-1112 inhibited STAT3 activation and its downstream signals, including Bcl-xL and cyclin D1. In human PC cell lines, P-V enhanced the growth inhibitory effect of gemcitabine (GEM), Abraxane and 5-FU, but not that of irinotecan. Normal human pancreatic epithelial cells were more resistant to the cytotoxic effects of MDC-1112/GEM combination. Furthermore, MDC-1112 enhanced GEM's effect on colony formation, apoptosis, cell migration, and cell invasion. In vivo, MDC-1112 and GEM, given alone, reduced patient-derived pancreatic tumor xenograft growth by 58% and 87%, respectively; whereas MDC-1112/GEM combination reduced tumor growth by 94%, inducing tumor stasis. In conclusion, MDC-1112 should be further explored as a potential agent to be used in combination with GEM for treating PC.


Assuntos
Anormalidades Múltiplas/tratamento farmacológico , Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Ceratocone/congênito , Organofosfatos/farmacologia , Neoplasias Pancreáticas/tratamento farmacológico , Ácido Valproico/análogos & derivados , Anormalidades Múltiplas/patologia , Animais , Apoptose/efeitos dos fármacos , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Desoxicitidina/farmacologia , Modelos Animais de Doenças , Humanos , Ceratocone/tratamento farmacológico , Ceratocone/patologia , Camundongos , Neoplasias Pancreáticas/patologia , Transdução de Sinais/efeitos dos fármacos , Ácido Valproico/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto , Gencitabina
15.
Nanomedicine (Lond) ; 14(21): 2873-2889, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31735124

RESUMO

Nucleic acid-based therapeutics are evaluated for their potential of treating a plethora of diseases, including cancer and inflammation. Short nucleic acids, such as miRNAs, have emerged as versatile regulators for gene expression and are studied for therapeutic purposes. However, their inherent instability in vivo following enteral and parenteral administration has prompted the development of novel methodologies for their delivery. Although research on the oral delivery of siRNAs is progressing, with the development and utilization of promising carrier-based methodologies for the treatment of a plethora of gastrointestinal diseases, research on miRNA-based oral therapeutics is lagging behind. In this review, we present the potential role of miRNAs in diseases of the GI tract, and analyze current research and the cardinal features of the novel carrier systems used for nucleic acid oral delivery that can be expanded for oral miRNA administration.


Assuntos
Gastroenteropatias/terapia , MicroRNAs/química , Nanocápsulas/química , Administração Oral , Animais , Transporte Biológico , Trato Gastrointestinal/efeitos dos fármacos , Regulação da Expressão Gênica , Técnicas de Transferência de Genes , Terapia Genética/métodos , Humanos , Lipídeos/química , MicroRNAs/metabolismo , Polímeros/química , RNA Interferente Pequeno/química , RNA Interferente Pequeno/metabolismo , Propriedades de Superfície
16.
Cancers (Basel) ; 11(10)2019 Oct 05.
Artigo em Inglês | MEDLINE | ID: mdl-31590367

RESUMO

Pancreatic cancer is a complex disease, in need of new therapeutic approaches. In this study, we explored the effect and mechanism of action of epigallocatechin-3-gallate (EGCG), a major polyphenol in green tea, alone and in combination with current chemotherapeutics on pancreatic cancer cell growth, focusing on glycolysis metabolism. Moreover, we investigated whether EGCG's effect is dependent on its ability to induce reactive oxygen species (ROS). EGCG reduced pancreatic cancer cell growth in a concentration-dependent manner and the growth inhibition effect was further enhanced under glucose deprivation conditions. Mechanistically, EGCG induced ROS levels concentration-dependently. EGCG affected glycolysis by suppressing the extracellular acidification rate through the reduction of the activity and levels of the glycolytic enzymes phosphofructokinase and pyruvate kinase. Cotreatment with catalase abrogated EGCG's effect on phosphofructokinase and pyruvate kinase. Furthermore, EGCG sensitized gemcitabine to inhibit pancreatic cancer cell growth in vitro and in vivo. EGCG and gemcitabine, given alone, reduced pancreatic tumor xenograft growth by 40% and 52%, respectively, whereas the EGCG/gemcitabine combination reduced tumor growth by 67%. EGCG enhanced gemcitabine's effect on apoptosis, cell proliferation, cell cycle and further suppressed phosphofructokinase and pyruvate kinase levels. In conclusion, EGCG is a strong combination partner of gemcitabine reducing pancreatic cancer cell growth by suppressing glycolysis.

17.
Mol Cancer Ther ; 18(11): 1961-1972, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31439714

RESUMO

The goal of this study was to evaluate combination of a novel taxoid, DHA-SBT-1214 chemotherapy, in modulating immune checkpoint marker expression and ultimately in improving antibody-based checkpoint blockade therapy in pancreatic adenocarcinoma (PDAC). DHA-SBT-1214 was encapsulated in an oil-in-water nanoemulsion and administered systemically in Panc02 syngeneic PDAC-bearing C57BL/6 mice. Following treatment with DHA-SBT-1214, expression levels of PD-L1 were measured and anti-PD-L1 antibody was administered in combination. The effects of combination therapy on efficacy and the molecular basis of synergistic effects were evaluated. PD-L1 expression was lower on Panc02 pancreatic tumor cells in vitro, which significantly increased after exposure to different chemotherapy drugs. Administration of DHA-SBT-1214, gemcitabine, and PD-L1 antibody alone failed to increase CD8+ T-cell infiltration inside tumors. However, combination of anti-PD-L1 therapy with a novel chemotherapy drug DHA-SBT-1214 in nanoemulsion (NE-DHA-SBT-1214) significantly enhanced CD8+ T-cell infiltration and the therapeutic effects of the anti-PD-L1 antibody. Furthermore, in the Panc02 syngeneic model, the NE-DHA-SBT-1214 combination therapy group reduced tumor growth to a higher extend than paclitaxel, nab-paclitaxel (Abraxane), gemcitabine, or single anti-PD-L1 antibody therapy groups. Our results indicate that NE-DHA-SBT-1214 stimulated immunogenic potential of PDAC and provided an enhanced therapeutic effect with immune checkpoint blockade therapy, which warrants further evaluation.


Assuntos
Antineoplásicos Imunológicos/administração & dosagem , Antígeno B7-H1/metabolismo , Carcinoma Ductal Pancreático/tratamento farmacológico , Ácidos Docosa-Hexaenoicos/química , Neoplasias Pancreáticas/tratamento farmacológico , Taxoides/administração & dosagem , Animais , Antineoplásicos Imunológicos/farmacologia , Antígeno B7-H1/genética , Carcinoma Ductal Pancreático/genética , Carcinoma Ductal Pancreático/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Composição de Medicamentos , Sinergismo Farmacológico , Emulsões , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Linfócitos do Interstício Tumoral/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Taxoides/química , Taxoides/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
18.
Nutrients ; 11(8)2019 Aug 09.
Artigo em Inglês | MEDLINE | ID: mdl-31405071

RESUMO

Most pancreatic cancers are usually diagnosed at an advanced stage when they have already metastasized. Epigallocatechin-3-gallate (EGCG), a major polyphenolic constituent of green tea, has been shown to reduce pancreatic cancer growth, but its effect on metastasis remains elusive. This study evaluated the capacity of EGCG to inhibit pancreatic cancer cell migration and invasion and the underlying mechanisms. EGCG reduced pancreatic cancer cell growth, migration, and invasion in vitro and in vivo. EGCG prevented "Cadherin switch" and decreased the expression level of TCF8/ZEB1, ß-Catenin, and Vimentin. Mechanistically, EGCG inhibited the Akt pathway in a time-dependent manner, by suppressing IGFR phosphorylation and inducing Akt degradation. Co-treatment with catalase or N-Acetyl-L-cysteine did not abrogate EGCG's effect on the Akt pathway or cell growth. Moreover, EGCG synergized with gemcitabine to suppress pancreatic cancer cell growth, migration, and invasion, through modulating epithelial-mesenchymal transition markers and inhibiting Akt pathway. In summary, EGCG may prove beneficial to improve gemcitabine sensitivity in inhibiting pancreatic cancer cell migration and invasion, to some extent through the inhibition of Akt pathway and epithelial-mesenchymal transition.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/farmacologia , Catequina/análogos & derivados , Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Desoxicitidina/análogos & derivados , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Neoplasias Pancreáticas/tratamento farmacológico , Proteínas Proto-Oncogênicas c-akt/metabolismo , Animais , Catequina/farmacologia , Linhagem Celular Tumoral , Desoxicitidina/farmacologia , Estabilidade Enzimática , Humanos , Camundongos Endogâmicos C57BL , Invasividade Neoplásica , Neoplasias Pancreáticas/enzimologia , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/patologia , Proteólise , Proteínas Proto-Oncogênicas c-akt/genética , Transdução de Sinais , Carga Tumoral , Gencitabina
19.
Carcinogenesis ; 40(12): 1480-1491, 2019 12 31.
Artigo em Inglês | MEDLINE | ID: mdl-30994173

RESUMO

New therapeutic strategies against glioblastoma multiforme (GBM) are urgently needed. Signal transducer and activator of transcription 3 (STAT3), constitutively active in many GBM tumors, plays a major role in GBM tumor growth and represents a potential therapeutic target. We have documented previously that phospho-valproic acid (MDC-1112), which inhibits STAT3 activation, possesses strong anticancer properties in multiple cancer types. In this study, we explored the anticancer efficacy of MDC-1112 in preclinical models of GBM, and evaluated its mode of action. MDC-1112 inhibited the growth of multiple human GBM cell lines in a concentration- and time-dependent manner. Normal human astrocytes were resistant to MDC-1112, indicating selectivity. In vivo, MDC-1112 reduced the growth of subcutaneous GBM xenografts in mice by up to 78.2% (P < 0.01), compared with the controls. Moreover, MDC-1112 extended survival in an intracranial xenograft model. Although all vehicle-treated mice died by 19 days of treatment, 7 of 11 MDC-1112-treated mice were alive and healthy by the end of 5 weeks, with many showing tumor regression. Mechanistically, MDC-1112 inhibited STAT3 phosphorylation at the serine 727 residue, but not at tyrosine 705, in vitro and in vivo. STAT3 overexpression rescued GBM cells from the cell growth inhibition by MDC-1112. In addition, MDC-1112 reduced STAT3 levels in the mitochondria and enhanced mitochondrial levels of reactive oxygen species, which triggered apoptosis. In conclusion, MDC-1112 displays strong efficacy in preclinical models of GBM, with the serine 727 residue of STAT3 being its key molecular target. MDC-1112 merits further evaluation as a drug candidate for GBM. New therapeutic options are needed for glioblastoma. The novel agent MDC-1112 is an effective anticancer agent in multiple animal models of glioblastoma, and its mechanism of action involves the inhibition of STAT3 phosphorylation, primarily at its Serine 727 residue.


Assuntos
Antineoplásicos/farmacologia , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Organofosfatos/farmacologia , Fator de Transcrição STAT3/metabolismo , Ácido Valproico/análogos & derivados , Animais , Neoplasias Encefálicas/metabolismo , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Feminino , Glioblastoma/metabolismo , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT3/efeitos dos fármacos , Ácido Valproico/farmacologia , Ensaios Antitumorais Modelo de Xenoenxerto
20.
J Natl Cancer Inst ; 111(2): 137-145, 2019 02 01.
Artigo em Inglês | MEDLINE | ID: mdl-29860330

RESUMO

BACKGROUND: Aspirin use is associated with reduced risk of several cancers. A pooled analysis of 12 case-control studies showed a 10% decrease in ovarian cancer risk with regular aspirin use, which was stronger for daily and low-dose users. To prospectively investigate associations of analgesic use with ovarian cancer, we analyzed data from 13 studies in the Ovarian Cancer Cohort Consortium (OC3). METHODS: The current study included 758 829 women who at study enrollment self-reported analgesic use, among whom 3514 developed ovarian cancer. Using Cox regression, we assessed associations between frequent medication use and risk of ovarian cancer. Dose and duration were also evaluated. All statistical tests were two-sided. RESULTS: Women who used aspirin almost daily (≥6 days/wk) vs infrequent/nonuse experienced a 10% reduction in ovarian cancer risk (rate ratio [RR] = 0.90, 95% confidence interval [CI] = 0.82 to 1.00, P = .05). Frequent use (≥4 days/wk) of aspirin (RR = 0.95, 95% CI = 0.88 to 1.03), nonaspirin nonsteroidal anti-inflammatory drugs (NSAIDs; RR = 1.00, 95% CI = 0.90 to 1.11), or acetaminophen (RR = 1.05, 95% CI = 0.88 to 1.24) was not associated with risk. Daily acetaminophen use (RR = 1.28, 95% CI = 1.00 to 1.65, P = .05) was associated with elevated ovarian cancer risk. Risk estimates for frequent, long-term (10+ years) use of aspirin (RR = 1.15, 95% CI = 0.98 to 1.34) or nonaspirin NSAIDs (RR = 1.19, 95% CI = 0.84 to 1.68) were modestly elevated, although not statistically significantly so. CONCLUSIONS: This large, prospective analysis suggests that women who use aspirin daily have a slightly lower risk of developing ovarian cancer (∼10% lower than infrequent/nonuse)-similar to the risk reduction observed in case-control analyses. The observed potential elevated risks for 10+ years of frequent aspirin and NSAID use require further study but could be due to confounding by medical indications for use or variation in drug dosing.


Assuntos
Analgésicos/uso terapêutico , Anti-Inflamatórios não Esteroides/uso terapêutico , Aspirina/uso terapêutico , Neoplasias Ovarianas/prevenção & controle , Medição de Risco/métodos , Europa (Continente)/epidemiologia , Feminino , Seguimentos , Humanos , Pessoa de Meia-Idade , Neoplasias Ovarianas/epidemiologia , Prognóstico , Estudos Prospectivos , Fatores de Risco , Estados Unidos/epidemiologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...