Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 29
Filtrar
1.
Int J Cancer ; 142(12): 2599-2609, 2018 06 15.
Artigo em Inglês | MEDLINE | ID: mdl-29388200

RESUMO

Natural killer (NK) cells exhibit strong cytotoxic activity against tumor cells without prior sensitization, and have the potential to exert antibody-dependent cellular cytotoxicity (ADCC). In this clinical trial, we examined the safety and efficacy of the use of NK cells, generated using a novel expansion system, in combination with IgG1 antibodies for the treatment of advanced gastric or colorectal cancers. Treatment consisted of trastuzumab- or cetuximab-based chemotherapy, plus adoptive NK cell therapy. For administration of expanded NK cells, dose escalation with a sequential 3 + 3 design was performed in three steps, at doses of 0.5 × 109 , 1.0 × 109 , and 2.0 × 109 cells/injection (N = 9). After 3 days of IgG1 antibody administration, patients were infused with expanded NK cells three times at triweekly intervals. NK cell populations expanded with our system were confirmed as being enriched in NK cells (median 92.9%) with high expression of NKG2D (97.6%) and CD16 (69.6%). The combination therapy was very well tolerated with no severe adverse events. Among six evaluable patients, four presented stable disease (SD) and two presented progressive disease. Of the four SD patients, three showed an overall decrease in tumor size after combination therapy. Immune monitoring suggested that combination therapy enhanced whole blood IFN-γ production and reduced peripheral regulatory T cells (Tregs). In conclusion, this phase I trial provides evidence of good tolerability, induction of Th1 immune responses, and preliminary anti-tumor activity for this combination therapy, in patients with advanced gastric and colorectal cancer that have received previous therapy.


Assuntos
Neoplasias Colorretais/terapia , Imunoglobulina G/uso terapêutico , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/transplante , Neoplasias Gástricas/terapia , Adulto , Idoso , Anticorpos Monoclonais Humanizados/uso terapêutico , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Cetuximab/uso terapêutico , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Trastuzumab/uso terapêutico
2.
Int J Cancer ; 137(11): 2558-65, 2015 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-26016447

RESUMO

The phenotype and severity of cancer cachexia differ among tumor types and metastatic site in individual patients. In this study, we evaluated if differences in tumor microenvironment would affect the development of cancer cachexia in a murine model, and demonstrated that body weight, adipose tissue and gastrocnemius muscle decreased in tumor-bearing mice. Interestingly, a reduction in heart weight was observed in the intraperitoneal tumor group but not in the subcutaneous group. We evaluated 23 circulating cytokines and members of the TGF-ß family, and found that levels of IL-6, TNF-α and activin A increased in both groups of tumor-bearing mice. Eotaxin and G-CSF levels in the intraperitoneal tumor group were higher than in the subcutaneous group. Atrogin 1 and MuRF1 mRNA expressions in the gastrocnemius muscle increased significantly in both groups of tumor-bearing mice, however, in the myocardium, expression of these mRNAs increased in the intraperitoneal group but not in subcutaneous group. Based on these results, we believe that differences in microenvironment where tumor cells develop can affect the progression and phenotype of cancer cachexia through alterations in various circulating factors derived from the tumor microenvironment.


Assuntos
Caquexia/patologia , Músculo Esquelético/patologia , Atrofia Muscular/patologia , Ativinas/sangue , Animais , Caquexia/sangue , Caquexia/genética , Linhagem Celular Tumoral , Modelos Animais de Doenças , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Proteínas Musculares/genética , Atrofia Muscular/sangue , Atrofia Muscular/genética , Miocárdio/patologia , RNA Mensageiro/genética , Proteínas Ligases SKP Culina F-Box/genética , Fator de Crescimento Transformador beta/sangue , Proteínas com Motivo Tripartido , Microambiente Tumoral/genética , Fator de Necrose Tumoral alfa/sangue , Ubiquitina-Proteína Ligases/genética
3.
J Transl Med ; 13: 277, 2015 Aug 25.
Artigo em Inglês | MEDLINE | ID: mdl-26303618

RESUMO

BACKGROUND: NK cells can destroy tumor cells without prior sensitization or immunization. Tumors often lose expression of MHC molecules and/or antigens. However, NK cells can lyse tumor cells in a non-MHC-restricted manner and independent of the expression of tumor-associated antigens. NK cells are therefore considered ideal for adoptive cancer immunotherapy; however the difficulty of obtaining large numbers of fully functional NK cells that are safe to administer deters its clinical use. This phase I clinical trial seeks to address this obstacle by first developing a novel system that expands large numbers of highly activated clinical grade NK cells, and second, determining if these cells are safe in a mono-treatment so they can be combined with other reagents in the next round of clinical trials. METHODS: Patients with unresectable, locally advanced and/or metastatic digestive cancer who did not succeed with standard therapy were enrolled. NK cells were expanded ex vivo by stimulating PBMCs with OK432, IL-2, and modified FN-CH296 induced T cells. Patients were administered autologous natural killer cell three times weekly via intravenous infusions in a dose-escalating manner (dose 0.5 × 10(9), 1.0 × 10(9), 2.0 × 10(9) cells/injection, three patients/one cohort). RESULTS: Total cell population had a median expansion of 586-fold (range 95-1102), with a significantly pure (90.96 %) NK cell population. Consequently, NK cells were expanded to approximately 4720-fold (range 1372-14,116) with cells being highly lytic in vitro and strongly expressing functional markers such as NKG2D and CD16. This NK cell therapy was very well tolerated with no severe adverse events. Although no clinical responses were observed, cytotoxicity of peripheral blood was elevated approximately twofolds up to 4 weeks post the last transfer. CONCLUSION: We successfully generated large numbers of activated NK cells from small quantities of blood without prior purification of the cells. We also determined that the expanded cells were safe to administer in a monotherapy and are suitable for the next round of clinical trials where their efficacy will be tested combined with other reagents. TRIAL REGISTRATION: UMIN UMIN000007527.


Assuntos
Neoplasias Gastrointestinais/terapia , Imunoterapia , Células Matadoras Naturais/citologia , Idoso , Feminino , Humanos , Imunofenotipagem , Células Matadoras Naturais/imunologia , Masculino , Pessoa de Meia-Idade
4.
J Clin Biochem Nutr ; 55(1): 56-61, 2014 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25120280

RESUMO

Epithelial-mesenchymal transition (EMT) plays a crucial role in cancer metastasis. In this study, we evaluated the effect of heat treatment on tumor growth factor-ß1 (TGF-ß1)-induced EMT in pancreatic cancer cells and tried to ascertain the mechanism related to any observed effects. Human pancreatic cancer cell lines (BxPC-3, PANC-1 and MIAPaCa-2) were stimulated by TGF-ß1, and evaluated for morphological changes using immunofluorescence and EMT-related factors (i.e., E-cadherin, Vimentin, Snail or ZEB-1) using RT-PCR. To examine the effect of heat on EMT, the cancer cells were heat-treated at 43°C for 1 h then stimulated with TGF-ß1. We then evaluated whether or not heat treatment changed the expression of EMT-related factors and cell migration and also whether Smad activation was inhibited in TGF-ß signaling. After being treated with TGF-ß1, pancreatic cancer cells resulted in EMT and cell migration was enhanced. Heat treatment inhibited TGF-ß1-induced changes in morphology, inhibited the expression of EMT-related factors, and attenuated TGF-ß1-induced migration in pancreatic cancer cells. Additionally, we observed that heat treatment blocked TGF-ß1-induced phosphorylation of Smad2 in PANC-1 cells. Our results suggest that heat treatment can suppress TGF-ß1-induced EMT and opens the possibility of a new therapeutic use of hyperthermia as a potential treatment for cancer metastasis.

5.
Int J Cancer ; 133(5): 1119-25, 2013 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-23420507

RESUMO

A core challenge in administering immune-based treatments for cancer is the establishment of easily accessible immunological assays that can predict patients' clinical responses to immunotherapy. In this study, our aim was to predict the therapeutic effects of adoptive T-cell therapy in patients with advanced pancreatic cancer. To do this, we evaluated whole blood cytokine levels and peripheral regulatory T cells (Tregs) in 46 patients with unresectable or recurrent pancreatic cancer who received adoptive T-cell therapy at 2-week intervals. To test immune function, venous blood was obtained from patients before the start of therapy and 2 weeks after the 4th treatment. Whole blood interferon (IFN)-α levels (after stimulation with the Sendai virus) were evaluated, as well as the levels of 9 cytokines stimulated with phytohemagglutinin [interleukin (IL)-2, IL-4, IL-5, IL-10, IL-12(p70), IL-13, tumor necrosis factor-α, IFN-γ, and granulocyte-monocyte colony-stimulating factor]. Peripheral Tregs were analyzed by flow cytometry. Using the obtained data, we then observed the relationship between these immunological parameters and clinical outcome of patients. We found that the whole blood production of IFN-γ, IL-2, IL-4, IL-5 and IL-13 significantly increased after adoptive T-cell therapy, whereas the number of peripheral Tregs did not change. Multivariate Cox proportional hazards analyses indicated that the number of peripheral Tregs before receiving adoptive T-cell therapy and the change in IFN-γ levels after adoptive T-cell therapy were independent variables predicting overall survival. The findings of this study indicate that the assay of whole blood IFN-γ production offers promise for evaluating the clinical response of patients to cancer immunotherapy.


Assuntos
Imunoterapia Adotiva , Interferon gama/sangue , Neoplasias Pancreáticas/terapia , Linfócitos T/imunologia , Adulto , Idoso , Idoso de 80 Anos ou mais , Citocinas/sangue , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/imunologia , Neoplasias Pancreáticas/mortalidade , Modelos de Riscos Proporcionais
6.
Int J Hyperthermia ; 28(7): 597-604, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22838644

RESUMO

PURPOSE: Despite advances in cancer therapy, treating pancreatic cancer remains one of the major challenges in the field of medical oncology. We conducted this phase II study to evaluate the efficacy and safety of regional hyperthermia combined with gemcitabine for the treatment of unresectable advanced pancreatic cancer. METHODS: Eligibility criteria included histologically proven, locally advanced or metastatic pancreatic cancer. Gemcitabine was administered intravenously at a dose of 1000 mg/m(2) on days 1, 8, and 15 every 4 weeks. Regional hyperthermia was performed once weekly, 1 day preceding or following gemcitabine administration. The primary end point was the 1-year survival rate. Secondary objectives were determination of tumour response and safety. RESULTS: We enrolled 18 patients with advanced pancreatic cancer between November 2008 and May 2010. The major grade 3-4 adverse events were neutropenia and anaemia; however, there were no episodes of infection. The objective response rate (ORR) and disease control rate (ORR + stable disease) were 11.1% and 61.1%, respectively. Median overall survival (OS) was 8 months, and the 1-year survival rate was 33.3%. Median OS of patients with locally advanced pancreatic cancer was 17.7 months. CONCLUSIONS: Regional hyperthermia combined with gemcitabine is well tolerated and active in patients with locally advanced pancreatic cancer.


Assuntos
Antimetabólitos Antineoplásicos/administração & dosagem , Desoxicitidina/análogos & derivados , Hipertermia Induzida , Neoplasias Pancreáticas/terapia , Idoso , Antimetabólitos Antineoplásicos/efeitos adversos , Terapia Combinada , Desoxicitidina/administração & dosagem , Desoxicitidina/efeitos adversos , Feminino , Humanos , Hipertermia Induzida/efeitos adversos , Masculino , Pessoa de Meia-Idade , Neoplasias Pancreáticas/mortalidade , Taxa de Sobrevida , Gencitabina
7.
Oncol Res ; 19(12): 543-54, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-22812187

RESUMO

We examined the effects of adoptive T-cell transfer (ACT) on the population of regulatory T cells (Tregs) in a mouse colorectal cancer transplant model. In an in vivo study, Treg populations in Balb/c mice colon26 transplant model after ACT were analyzed in peripheral blood, local lymph node, and tumor. In an in vitro study CD4+ cells were cultured in medium containing TGF-beta to induce Tregs. LAK cells were added or not in this Treg induction system. Treg induction after coculture with LAK was investigated. We also studied the role of IFN-gamma in the mechanism of Treg induction. Tregs in the draining lymph nodes and tumor were significantly suppressed by ACT. The induction of Tregs in vitro was inhibited by coculture with LAK cells. Furthermore, Tregs in the cultured cells were significantly inhibited by addition of exogenous IFN-gamma. Moreover, Tregs were increased by addition of IFN-gamma mAb. ACT may decrease Tregs in tumor-bearing hosts. One of the mechanisms is considered to be IFN-gamma inhibiting the induction of Tregs.


Assuntos
Adenocarcinoma/imunologia , Linfócitos T CD4-Positivos/transplante , Neoplasias do Colo/imunologia , Modelos Animais de Doenças , Imunoterapia , Células Matadoras Ativadas por Linfocina/transplante , Linfócitos T Reguladores/imunologia , Adenocarcinoma/prevenção & controle , Transferência Adotiva , Animais , Western Blotting , Linfócitos T CD4-Positivos/imunologia , Neoplasias do Colo/prevenção & controle , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Fatores de Transcrição Forkhead , Técnicas Imunoenzimáticas , Interferon gama/metabolismo , Células Matadoras Ativadas por Linfocina/imunologia , Linfonodos/imunologia , Linfonodos/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Linfócitos T Reguladores/citologia , Fator de Crescimento Transformador beta/farmacologia , Células Tumorais Cultivadas
8.
Chemistry ; 16(26): 7750-9, 2010 Jul 12.
Artigo em Inglês | MEDLINE | ID: mdl-20564294

RESUMO

Core-shell-structured nanoparticles, consisting of a noble metal or metal oxide core and a chromia (Cr(2)O(3)) shell, were studied as promoters for photocatalytic water splitting under visible light. Core nanoparticles were loaded by impregnation, adsorption or photodeposition onto a solid solution of gallium nitride and zinc oxide (abbreviated GaN:ZnO), which is a particulate semiconductor photocatalyst with a band gap of approximately 2.7 eV, and a Cr(2)O(3) shell was formed by photodeposition using a K(2)CrO(4) precursor. Photodeposition of Cr(2)O(3) on GaN:ZnO modified with a noble metal (Rh, Pd and Pt) or metal oxide (NiO(x), RuO(2) and Rh(2)O(3)) co-catalyst resulted in enhanced photocatalytic activity for overall water splitting under visible light (lambda>400 nm). This enhancement in activity was primarily due to the suppression of undesirable reverse reactions (H(2)-O(2) recombination and/or O(2) photoreduction) and/or protection of the core component from chemical corrosion, depending on the core type. Among the core materials examined, Rh species exhibited relatively high performance for this application. The activity for visible-light water splitting on GaN:ZnO modified with an Rh/Cr(2)O(3) core-shell configuration was dependent on both the dispersion of Rh nanoparticles and the valence state. In addition, the morphology of the Cr(2)O(3) photodeposits was significantly affected by the valence state of Rh and the pH at which the photoreduction of K(2)CrO(4) was conducted. When a sufficient amount of K(2)CrO(4) was used as the precursor and the solution pH ranged from 3 to 7.5, Cr(2)O(3) was successfully formed with a constant shell thickness (approximately 2 nm) on metallic Rh nanoparticles, which resulted in an effective promoter for overall water splitting.

9.
Oncol Rep ; 44(1): 371-381, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32627027

RESUMO

Epithelial­mesenchymal transition (EMT) is considered a crucial event in the development of cancer metastasis. Metformin is a drug used in the treatment of type 2 diabetes. Recently, increasing evidence has indicated that metformin possesses anti­tumor activities. However, the effects of metformin on EMT and metastases in pancreatic cancer remain unknown. Thus, the present study investigated whether metformin inhibits EMT of human pancreatic cancer cell lines. Pancreatic cancer cells were stimulated with transforming growth factor ß1 (TGF­ß1), an activator of EMT signaling, with or without metformin. After 48 h, the levels of epithelial and mesenchymal markers were evaluated by western blot analysis, immunocytochemistry and RT­qPCR. Cancer cell migration was evaluated by an in vitro wound healing assay. The cells stimulated with TGF­ß1 acquired an elongated and fusiform morphology, which was inhibited by metformin. The wound healing assay revealed that metformin significantly suppressed the TGF­ß1­stimulated migration of pancreatic cancer cells. Following treatment with metformin, E­cadherin expression (epithelial marker) was upregulated, and the levels of mesenchymal markers were downregulated, which had been increased by TGF­ß1 in these cells. Exposure of the cells to TGF­ß1 activated the Smad2/3 and Akt/mammalian target of rapamycin (mTOR) pathways, and this effect was inhibited by metformin, suggesting that metformin inhibits TGF­ß1­induced­EMT through the down­regulation of the Smad pathway in PANC­1 cells and the downregulation of the Akt/mTOR pathway in BxPC­3 cells. In an animal model of surgical orthotopic implantation, metformin inhibited liver metastasis without a significant reduction in the size of the primary pancreatic tumor. On the whole, the findings of the present study suggest that metformin inhibits EMT and cancer metastasis through the Smad or Akt/mTOR pathway.


Assuntos
Antígenos CD/genética , Caderinas/genética , Neoplasias Hepáticas/tratamento farmacológico , Neoplasias Hepáticas/secundário , Metformina/administração & dosagem , Neoplasias Pancreáticas/tratamento farmacológico , Fator de Crescimento Transformador beta1/farmacologia , Animais , Linhagem Celular Tumoral , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Feminino , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Neoplasias Hepáticas/genética , Neoplasias Hepáticas/metabolismo , Metformina/farmacologia , Camundongos , Neoplasias Pancreáticas/genética , Neoplasias Pancreáticas/metabolismo , Transdução de Sinais/efeitos dos fármacos , Ensaios Antitumorais Modelo de Xenoenxerto
10.
Nihon Shokakibyo Gakkai Zasshi ; 106(4): 554-9, 2009 Apr.
Artigo em Japonês | MEDLINE | ID: mdl-19346725

RESUMO

A 58-year-old man was brought to our hospital with left upper abdominal pain which suddenly appeared on the previous evening. An abdominal CT scan showed localized retention of ascites, a slightly high density mass under the left upper abdominal wall, with a high density area detected within the mass, which was suggestive of leakage of contrast medium from peripheral branches of the omental artery. From these findings intraperitoneal hemorrhage caused by bleeding from the greater omentum was suspected. Angiographic examination of the abdomen indicated extravasation of contrast medium from blood vessels of the right gastroepiploic artery. Transarterial embolization was carried out and permanent hemostasis was achieved. Injury, anticoagulant, neoplasms, varix, torsion of the omentum, and segmental arterial mediolysis (SAM) etc have been reported as causes of omental bleeding, but none of these were found in our case. We diagnosed the present case as idiopathic omental bleeding.


Assuntos
Embolização Terapêutica/métodos , Hemorragia/terapia , Omento/irrigação sanguínea , Artéria Gastroepiploica , Humanos , Masculino , Pessoa de Meia-Idade
11.
Med Oncol ; 36(7): 60, 2019 May 27.
Artigo em Inglês | MEDLINE | ID: mdl-31134385

RESUMO

Immune checkpoint molecules are expressed on cancer cells and regulate tumor immunity by binding to ligands on immune cells. Although soluble forms of immune checkpoint molecules have been detected in the blood of patients with some types of tumors, their roles have not been fully elucidated. Soluble PD-L1, PD-1, CD155, LAG3, and CD226 (sPD-L1, sPD-1, sCD155, sLAG3, and sCD226, respectively) were measured in the sera of 47 patients with advanced esophageal cancer and compared with those of 24 control subjects. Pretreatment levels of sPD-1 and sCD155 were significantly higher in the patients with cancer than in the control subjects (P = 0.023, P = 0.001). The sPD-1 levels tended to be higher in the patients with lymph node metastasis, a large tumor diameter, and higher levels of serum SCC antigen (P = 0.150, P = 0.189, and P = 0.078, respectively). However, higher levels of sCD155 were associated with a better response to chemotherapy and favorable overall survival (P = 0.111 and P = 0.068, respectively). After 2 courses of chemotherapy, the levels of sCD155 and sCD226 were significantly increased (P < 0.001 and P = 0.002, respectively). Moreover, the increase in sCD226 during chemotherapy was associated with poor treatment response (P = 0.019). sPD-1 levels are possibly dependent on the tumor aggressiveness of the esophageal cancer. Furthermore, the pretreatment levels of sCD155 and kinetic change of sCD226 after chemotherapy may be used as biomarkers of the treatment response and prognosis in patients with esophageal cancer.


Assuntos
Protocolos de Quimioterapia Combinada Antineoplásica/uso terapêutico , Neoplasias Esofágicas/sangue , Neoplasias Esofágicas/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/sangue , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Adulto , Idoso de 80 Anos ou mais , Antígenos CD/sangue , Antígenos de Diferenciação de Linfócitos T/sangue , Antígeno B7-H1/sangue , Estudos de Casos e Controles , Cisplatino/administração & dosagem , Docetaxel/administração & dosagem , Neoplasias Esofágicas/imunologia , Carcinoma de Células Escamosas do Esôfago/imunologia , Feminino , Fluoruracila/administração & dosagem , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Receptor de Morte Celular Programada 1/sangue , Receptores Virais/sangue , Proteína do Gene 3 de Ativação de Linfócitos
12.
Med Oncol ; 36(2): 15, 2019 Jan 02.
Artigo em Inglês | MEDLINE | ID: mdl-30600347

RESUMO

Undernutrition and sarcopenia are associated with a higher incidence of chemotherapy-related toxicity and a poor prognosis in several kinds of cancer, but the impact of sarcopenia on the outcomes of chemotherapy for esophageal cancer remains unclear. Thus, the purpose of this retrospective study was to investigate whether sarcopenia affects the efficacy and toxicities of chemotherapy for advanced esophageal cancer patients. Data were collected from 31 esophageal cancer patients who underwent neo-adjuvant chemotherapy followed by surgery. Body composition was assessed at the start of chemotherapy by bioelectrical impedance analysis, and outcomes of chemotherapy were compared between sarcopenic and non-sarcopenic groups. Of the 31 patients, sarcopenia was observed in 16 (51.6%). The incidence of toxicities was not different between the two groups. However, as for pathologic response, a good therapeutic effect (Grade 2 or higher) was more common in the non-sarcopenic group than in the sarcopenic group (53.3% vs. 25.0%). Multivariate analysis showed that sarcopenia was an independent predictor of poor pathological response (odds ratio 8.02; P = 0.037). The results of this study suggest the potential utility of sarcopenia assessment in neoadjuvant patient selection strategies.


Assuntos
Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/patologia , Músculo Esquelético/patologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/patologia , Adulto , Idoso , Quimioterapia Adjuvante , Carcinoma de Células Escamosas do Esôfago/tratamento farmacológico , Carcinoma de Células Escamosas do Esôfago/patologia , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Terapia Neoadjuvante , Estadiamento de Neoplasias , Estudos Retrospectivos , Sarcopenia/patologia
13.
Oncol Rep ; 37(3): 1545-1554, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-28112370

RESUMO

Although improvements in the chemotherapy modalities for pancreatic cancer have been realized, pancreatic cancer remains one of the most lethal malignancies. New-generation cancer immunotherapy methods, such as blocking of the PD-1/PD-L1 pathway, are consistently being investigated to improve the survival of pancreatic cancer patients. In the present study, we evaluated the influence of anticancer agents 5-fluorouracil, gemcitabine and paclitaxel on PD-L1 expression in human pancreatic cancer cell lines MIA PaCa-2 and AsPC-1 and in murine pancreatic cancer cell line Pan02. Additionally, we analyzed the molecular mechanisms that facilitated the regulation of PD-L1 expression in these cell lines. We observed that when AsPC-1, MIA PaCa-2 and Pan02 cells were stimulated by 5-fluorouracil, gemcitabine or paclitaxel, PD-L1 surface protein expression was enhanced. Similarly, the mRNA level of PD-L1 was upregulated in the AsPC-1 and Pan02 cells when stimulated by each of the three anticancer agents. The phosphorylation of STAT1 and an increase in total STAT1 were also observed in the AsPC-1 cells when stimulated by each anticancer agent. In response to JAK2 inhibitor treatment, PD-L1 upregulation induced by the anticancer agents was reduced in a dose-dependent manner. These results suggest that i) the JAK2/STAT1 pathway is involved in the anticancer agent-mediated PD-L1 transcription; and ii) the anticancer agents altered the tumor immune response which may induce tumor immune escape. These findings can have an influence on the design of treatments that combine chemotherapy and immunotherapy.


Assuntos
Antineoplásicos/farmacologia , Antígeno B7-H1/metabolismo , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Janus Quinase 2/metabolismo , Neoplasias Pancreáticas/metabolismo , Fator de Transcrição STAT1/metabolismo , Apoptose/efeitos dos fármacos , Antígeno B7-H1/genética , Western Blotting , Proliferação de Células/efeitos dos fármacos , Citometria de Fluxo , Humanos , Janus Quinase 2/genética , Neoplasias Pancreáticas/tratamento farmacológico , Neoplasias Pancreáticas/patologia , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT1/genética , Transdução de Sinais , Células Tumorais Cultivadas
14.
Oncol Rep ; 36(2): 1093-100, 2016 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-27374272

RESUMO

Chemo (chemoradio) therapy can induce oral mucositis and change body composition in patients with esophageal cancer. The impact of the amino acid-rich elemental diet Elental® on oral mucositis and changes in body composition during chemo (chemoradio) therapy is unclear. Thus, the purpose of the present study was to examine the preventive effects of Elental on oral mucositis and sarcopenia progression during chemo (chemoradio) therapy for esophageal cancer. Patients were randomized to receive either azulene oral rinse (Arm 1) or Elental (Arm 2) during the treatment cycle (4 weeks). The incidence of oral mucositis and other adverse events was evaluated weekly. Body composition pre- and post-treatment cycle was measured by bioelectric impedance analysis. Thirty­three patients (17 azulene and 16 Elental) completed the study, and the groups were well matched. Elental tended to reduce the incidence of oral mucositis (Arm 1, 23.5% and Arm 2, 12.5%), but there was no statistically significant difference between the groups. The average body mass index (BMI) and body fat mass decreased significantly in both groups after the treatments. Lean body mass (LBM) was reduced in Arm 1, but was increased in Arm 2 after the treatment; the relative change of LBM after the treatment was significant between Arm 1 and Arm 2 (P=0.007). This study revealed that Elental nutrition could counteract sarcopenia development during chemoradiotherapy for esophageal cancer. These properties may lead to improvement of the quality of life and clinical outcome of esophageal cancer patients treated with chemo (chemoradio) therapy (Clinical Trial Registry ID: UMIN 000007960).


Assuntos
Aminoácidos/administração & dosagem , Antineoplásicos/efeitos adversos , Antineoplásicos/uso terapêutico , Composição Corporal/efeitos dos fármacos , Quimiorradioterapia/efeitos adversos , Neoplasias Esofágicas/tratamento farmacológico , Neoplasias Esofágicas/radioterapia , Adulto , Idoso , Índice de Massa Corporal , Feminino , Alimentos Formulados , Humanos , Masculino , Pessoa de Meia-Idade , Qualidade de Vida , Estomatite/induzido quimicamente , Estomatite/prevenção & controle , Magreza
15.
Cancer Lett ; 229(2): 223-33, 2005 Nov 18.
Artigo em Inglês | MEDLINE | ID: mdl-16095811

RESUMO

The transcription factor NF-kappaB is reportedly activated by anti-cancer chemotherapeutic compounds in many cancer cell lines and NF-kappaB activation is one mechanism by which tumors become resistant to apoptosis. Antioxidants have been reported to serve as potent NF-kB inhibitors. In this study, we investigated the ability of edaravone to enhance apoptosis induced by CPT-11 through inhibition of NF-kB. In vitro, SN38, the active metabolite of CPT-11, induced activation of NF-kB, the production of intracellular reactive oxygen species, the activation of caspase-3, and apoptosis in colon26 cells. Pretreatment with edaravone scavenged the SN38-produced reactive oxygen species, and inhibited the SN38-induced activation of NF-kB. Moreover, edaravone enhanced the activation of caspase-3, and the level of apoptosis induced by SN38. In vivo, the combination of edaravone with CPT-11 reduced subcutaneous tumor growth and number of pulmonary metastases more effectively than CPT-11 alone. These results demonstrate that the combination of edaravone with CPT-11 may constitute a new strategy for treating primary and metastatic colon cancer.


Assuntos
Antineoplásicos/farmacologia , Protocolos de Quimioterapia Combinada Antineoplásica/administração & dosagem , Antipirina/análogos & derivados , Neoplasias do Colo/tratamento farmacológico , NF-kappa B/efeitos dos fármacos , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/secundário , Animais , Antineoplásicos/metabolismo , Antipirina/administração & dosagem , Apoptose/efeitos dos fármacos , Western Blotting , Camptotecina/administração & dosagem , Camptotecina/análogos & derivados , Camptotecina/metabolismo , Caspase 3 , Caspases/efeitos dos fármacos , Caspases/metabolismo , Linhagem Celular Tumoral , Neoplasias do Colo/patologia , Modelos Animais de Doenças , Edaravone , Ensaio de Desvio de Mobilidade Eletroforética , Sequestradores de Radicais Livres/administração & dosagem , Marcação In Situ das Extremidades Cortadas , Irinotecano , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/secundário , Masculino , Camundongos , NF-kappa B/metabolismo , Espécies Reativas de Oxigênio
16.
J Agric Food Chem ; 53(3): 562-5, 2005 Feb 09.
Artigo em Inglês | MEDLINE | ID: mdl-15686402

RESUMO

Six azaphilones, monascin (1), ankaflavin (2), rubropunctatin (3), monascorburin (4), rubropunctamine (5), and monascorburamine (6), two furanoisophthalides, xanthomonasin A (7) and xanthomonasin B (8), and two amino acids, (+)-monascumic acid (9) and (-)-monascumic acid (10), isolated from the extracts of Monascus pilosus-fermented rice (red-mold rice) were evaluated for their inhibitory effects on 12-O-tetradecanoylphorbol-13-acetate (TPA)-induced inflammation in mice, on the induction of Epstein-Barr virus early antigen (EBV-EA) by TPA in Raji cells, and on the activation of (+/-)-(E)-methyl-2[(E)-hydroxy-imino]-5-nitro-6-methoxy-3-hexemide (NOR 1), a nitric oxide (NO) donor. Among the compounds tested, seven compounds (1-6 and 10) on TPA-induced inflammation, and six compounds (1, 3-5, 9, and 10) on EBV-EA activation, exhibited potent inhibitory effects. All of the compounds tested showed moderate inhibitory effects on NOR 1 activation.


Assuntos
Benzopiranos/isolamento & purificação , Ácidos Dicarboxílicos/isolamento & purificação , Fermentação , Monascus/metabolismo , Oryza/química , Pigmentos Biológicos/isolamento & purificação , Xantonas/isolamento & purificação , Animais , Antígenos Virais/efeitos dos fármacos , Benzoatos/antagonistas & inibidores , Benzopiranos/uso terapêutico , Ácidos Dicarboxílicos/uso terapêutico , Feminino , Imidazóis/antagonistas & inibidores , Inflamação/prevenção & controle , Camundongos , Camundongos Endogâmicos ICR , Oryza/microbiologia , Pigmentos Biológicos/uso terapêutico , Xantonas/uso terapêutico
17.
Chem Biodivers ; 2(10): 1305-9, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17191930

RESUMO

Monascin (1) constitutes one of the azaphilonoid pigments in the extracts of Monascus pilosus-fermented rice (red-mold rice). Compound 1 was evaluated for its anti-tumor-initiating activity via oral administration on the two-stage carcinogenesis of mouse skin tumor induced by peroxynitrite (ONOO-; PN) or by ultraviolet light B (UVB) as an initiator and 12-O-tetradecanoylphorbol-13-acetate (TPA) as a promoter. Compound 1 exhibited marked inhibitory activity on both PN- and UVB-induced mouse skin carcinogenesis tests. These findings suggest that compound 1 may be valuable as potential cancer chemopreventive agent in chemical and environmental carcinogenesis.


Assuntos
Antineoplásicos/química , Antineoplásicos/farmacologia , Compostos Heterocíclicos com 3 Anéis/química , Compostos Heterocíclicos com 3 Anéis/farmacologia , Monascus/metabolismo , Oryza/metabolismo , Oryza/microbiologia , Neoplasias Cutâneas/patologia , Animais , Antineoplásicos/metabolismo , Feminino , Compostos Heterocíclicos com 3 Anéis/metabolismo , Camundongos , Camundongos Endogâmicos SENCAR , Estrutura Molecular , Extratos Vegetais/química , Extratos Vegetais/farmacologia , Neoplasias Cutâneas/induzido quimicamente , Organismos Livres de Patógenos Específicos , Fatores de Tempo
19.
Oncol Rep ; 33(5): 2545-52, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25738564

RESUMO

Although treatment with an antibody against cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) combined with multiple therapeutic interventions has been explored, the effect of combination therapy with CTLA-4 inhibition and adoptive T-cell therapy has not been determined. In the present study, our aim was to determine whether CTLA-4 inhibition, combined with adoptive transfer of T cells at different stages of differentiation, exhibits synergistic antitumor effects in a murine colon cancer model. Mice bearing subcutaneous tumors were administered adoptive T-cell transfer of CD62Lhigh or CD62Llow cells combined with an anti-CTLA-4 antibody (α-CTLA-4) or control immunoglobulin G. Subcutaneous tumors were harvested, and the antitumor effects and helper T-cell polarization were analyzed. CTLA-4 inhibition combined with CD62Lhigh cell administration showed the strongest antitumor effect. Combination therapy increased the number of CD3+ cells within the tumor. Moreover, CTLA-4 inhibition induced polarization of T cells infiltrating the tumor toward the T helper 1 lineage, and suppressed the frequency of regulatory T cells within the tumor, particularly in combination with CD62Lhigh T-cell transfer. This is the first report demonstrating that the efficacy of α-CTLA-4 and adoptive T-cell transfer combination therapy depends on the state of differentiation of the transferred T cells. Our data support the notion that a combination of α-CTLA-4 and adoptive T-cell transfer containing an abundance of naïve phenotype cells could potentially exert antitumor effects in a clinical setting.


Assuntos
Antineoplásicos/imunologia , Antígeno CTLA-4/imunologia , Linfócitos T Citotóxicos/imunologia , Linfócitos T Reguladores/imunologia , Transferência Adotiva/métodos , Animais , Antígenos CD/imunologia , Diferenciação Celular/imunologia , Linhagem Celular Tumoral , Terapia Baseada em Transplante de Células e Tecidos/métodos , Ativação Linfocitária/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C
20.
Life Sci ; 75(26): 3117-28, 2004 Nov 12.
Artigo em Inglês | MEDLINE | ID: mdl-15488892

RESUMO

Exercise has various beneficial effects on liver function, enhancing both nutrient metabolism and antioxidant capacity. To explore the molecular mechanisms underlying these changes, we used a high-density cDNA microarray containing probe sets for 2,845 genes to analyze changes of gene transcription in the livers of rats after 4 weeks of running exercise. In comparison with sedentary animals, 105 genes were up-regulated and 86 genes were down-regulated, including genes with unknown functions. In addition, we detected an increase of p38 mitogen-activated protein kinase and protein and of the protein for signal transducer and activator of transcription 3 (stat3), corresponding to the increase of these mRNAs shown by microarray analysis. These results indicate that long-term exercise can alter liver function via changes of gene expression, especially the genes encoding signal transduction proteins such as p38 and stat3.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Esforço Físico , Animais , Western Blotting , Primers do DNA , Proteínas de Ligação a DNA , Masculino , Análise de Sequência com Séries de Oligonucleotídeos , Ratos , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Fator de Transcrição STAT3 , Transdução de Sinais/fisiologia , Transativadores , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa