Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 25
Filter
1.
Gastric Cancer ; 20(Suppl 1): 128-134, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28028665

ABSTRACT

BACKGROUND: Despite recent progress in systemic chemotherapy, the prognosis of gastric cancer patients with peritoneal metastasis (P1) or positive peritoneal cytology findings (CY1) is still poor. We developed a regimen combining intraperitoneal (IP) paclitaxel (PTX) with S-1 and PTX, which can produce notable efficacy with regard to peritoneal lesions. Surgery after response to combination chemotherapy is a promising option for P1 or CY1 gastric cancer. A retrospective study was performed to evaluate the safety and efficacy. METHODS: This study enrolled 100 primary P1 or CY1 gastric cancer patients treated with IP PTX plus S-1 and PTX at the University of Tokyo Hospital between 2005 and 2011. Radical gastrectomy was performed when peritoneal cytology findings became negative, and the disappearance or obvious shrinkage of peritoneal metastasis was confirmed by laparoscopy. The same chemotherapy regimen was restarted after surgery and repeated with appropriate dose reduction. RESULTS: Gastrectomy was performed in 64 (P1 56, P0CY1 8) of 100 (P1 90, P0CY1 10) patients. R0 resection was achieved in 44 patients (69%). The median survival time was 30.5 months [95% confidence interval (CI) 23.6-37.7 months] from the initiation of intraperitoneal chemotherapy and 34.6 months (95% CI 26.8-39.4 months) from the diagnosis of gastric cancer. Postoperative complications included anastomotic leakage and pancreatic fistula, each in two patients, which were cured conservatively. There were no treatment-related deaths. The median survival time of the 36 patients who did not undergo surgery was 14.3 months (95% CI 10.0-17.8 months). CONCLUSIONS: Surgery after response to intraperitoneal and systemic chemotherapy is safe and may prolong the survival of P1 and CY1 gastric cancer patients.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Gastrectomy , Peritoneal Neoplasms/therapy , Stomach Neoplasms/therapy , Adult , Aged , Aged, 80 and over , Combined Modality Therapy , Drug Combinations , Female , Follow-Up Studies , Humans , Injections, Intraperitoneal , Lymphatic Metastasis , Male , Middle Aged , Neoplasm Staging , Oxonic Acid/administration & dosage , Paclitaxel/administration & dosage , Peritoneal Lavage , Peritoneal Neoplasms/secondary , Prognosis , Retrospective Studies , Stomach Neoplasms/pathology , Survival Rate , Tegafur/administration & dosage
2.
Gan To Kagaku Ryoho ; 39(7): 1127-30, 2012 Jul.
Article in Japanese | MEDLINE | ID: mdl-22790053

ABSTRACT

The patient was a 72-year-old woman, admitted to our hospital due to appetite loss. We performed gastroscopy, colonoscopy and abdominal CT. She had both advanced ascending colon cancer with multiple liver metastasis(cStage IV: cT3N1H3)and early gastric cancer(cStage 0: cTisN0M0). She received chemotherapy with modified FOLFOX6(mFOLFOX6), and the chemotherapy was judged effective for her gastric cancer. During the next 6 months, a total of 10 courses had been performed. The tumor marker scores(CEA, CA19-9)decreased significantly. Gastric cancer was diagnosed as CR(complete response)in gastroscopy after 6 courses of chemotherapy. In this case, chemotherapy with mFOLFOX6 was effective for gastric cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Colonic Neoplasms/drug therapy , Neoplasms, Second Primary/drug therapy , Stomach Neoplasms/drug therapy , Aged , Colonic Neoplasms/pathology , Female , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Neoplasm Staging , Neoplasms, Second Primary/pathology , Organoplatinum Compounds/therapeutic use , Stomach Neoplasms/pathology , Tomography, X-Ray Computed
3.
Eur J Gastroenterol Hepatol ; 22(2): 228-33, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923997

ABSTRACT

We report a male patient with double advanced tumors in the jejunum and descending colon and multiple lung tumors. The intestinal cancers were surgically resected. Immunoprofiling of the specimens revealed a rare phenotype: the jejunal cancer was positive for cytokeratin (CK) 7, partially positive for CK20, and Cdx-2-negative, whereas the colon cancer was CK7(+), CK20(-), and Cdx-2(-). Biopsied lung tumor was diagnosed as tubular adenocarcinoma, and CK7(+)/CK20(+)/Cdx-2(-). Together with clinical information, we deduced that the jejunal adenocarcinoma had presumably metastasized to the lung. Moreover, postoperative oxaliplatin, including chemotherapy, significantly reduced the lung metastases, suggesting that this regimen is a promising treatment option for advanced small bowel adenocarcinoma.


Subject(s)
Adenocarcinoma/secondary , Biomarkers, Tumor/analysis , Colonic Neoplasms/pathology , Immunohistochemistry , Jejunal Neoplasms/pathology , Lung Neoplasms/secondary , Neoplasms, Multiple Primary , Adenocarcinoma/chemistry , Adenocarcinoma/genetics , Adenocarcinoma/therapy , Antineoplastic Agents/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biopsy , CDX2 Transcription Factor , Chemotherapy, Adjuvant , Colectomy , Colonic Neoplasms/chemistry , Colonic Neoplasms/genetics , Colonic Neoplasms/therapy , Fatal Outcome , Fluorouracil/therapeutic use , Homeodomain Proteins/analysis , Humans , Jejunal Neoplasms/chemistry , Jejunal Neoplasms/genetics , Jejunal Neoplasms/therapy , Keratin-20/analysis , Keratin-7/analysis , Leucovorin/therapeutic use , Lung Neoplasms/chemistry , Lung Neoplasms/genetics , Lung Neoplasms/therapy , Lymph Node Excision , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/therapeutic use , Oxaliplatin , Phenotype , Tomography, X-Ray Computed , Treatment Outcome
4.
J Surg Res ; 155(1): 40-7, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19394652

ABSTRACT

Orally applicable Delta9-tetrahydrocannabinol and its synthetic derivatives have been used as antiemetic drugs during chemotherapy in cancer patients. However, it is not well known how cannabinoids influence the effects of chemotherapeutic agents on malignant tumors. In this study, we investigated how the endogenous cannabinoid anandamide (AEA) changes the effect of paclitaxel on gastric cancer cell lines. In the human gastric cancer cell line, HGC-27, which express cannabinoid receptor 1 (CB1), AEA stimulated proliferation at concentrations under 1 microM, while it strongly suppressed proliferation through the induction of apoptosis at 10 microM. This bimodal effect was reproduced by a selective CB1 agonist, arachidonyl-2-chloroethylamide, although the effects were less marked. When AEA was used with paclitaxel, AEA at 10 microM synergistically enhanced the cytotoxic effect of paclitaxel, whereas it showed no significant effect at lower concentrations. Flow cytometric analysis revealed that addition of 10 microM AEA synergistically enhanced paclitaxel-induced apoptosis, possibly through the activation of caspase-3, -8, and -9. Our results suggest that cannabinoids could be a good palliative agent for cancer patients receiving paclitaxel.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Arachidonic Acids/pharmacology , Cannabinoid Receptor Modulators/pharmacology , Paclitaxel/pharmacology , Polyunsaturated Alkamides/pharmacology , Stomach Neoplasms/drug therapy , Antineoplastic Agents, Phytogenic/therapeutic use , Apoptosis/drug effects , Arachidonic Acids/therapeutic use , Cannabinoid Receptor Modulators/therapeutic use , Caspases/drug effects , Caspases/metabolism , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Endocannabinoids , Enzyme Activation/drug effects , Humans , Paclitaxel/therapeutic use , Polyunsaturated Alkamides/therapeutic use , Receptor, Cannabinoid, CB1/antagonists & inhibitors , Receptor, Cannabinoid, CB1/metabolism , Receptor, Cannabinoid, CB2/antagonists & inhibitors , Receptor, Cannabinoid, CB2/metabolism
5.
J Surg Res ; 151(1): 115-20, 2009 Jan.
Article in English | MEDLINE | ID: mdl-18619615

ABSTRACT

BACKGROUND: Zoledronic acid (ZOL) is clinically available for the treatment of skeletal complications. In preclinical studies, strong anti-cancer activities against breast cancer, prostate cancer, and leukemia were reported. It also inhibited the proliferation of cultured human endothelial cells, suggestive of an anti-angiogenic activity. Since ZOL has the tendency to accumulate in bone, we investigated the effect of ZOL on endothelial progenitor cells (EPCs), which originate from the bone marrow, and play important roles in angiogenesis. MATERIALS AND METHODS: Human peripheral blood mononuclear cells were cultured for 7 d to differentiate into EPCs. Cells were treated without/with ZOL or with geranylgeraniol (GGOH). Their endothelial phenotype was confirmed by the expression of CD144 and vascular endothelial growth factor receptor 2 and the tube-like formation ability on Matrigel (Becton Dickinson, Bedford, MA). Annexin V/propidium iodide staining was used to analyze apoptosis. RESULTS: ZOL treatment, even at low doses, from d 2 to 7 of culture resulted in impaired EPC differentiation and could be restored by co-treatment with GGOH. On the other hand, treatment of putative EPCs with ZOL at concentrations higher than 10 mum resulted in induction of apoptosis. CONCLUSION: ZOL dose-dependently inhibited the differentiation of EPCs, the effect being observed even at low drug levels. At high concentrations, ZOL also induced the apoptotic death of putative EPCs. Since GGOH restored the inhibitory effect of ZOL on EPCs differentiation, the effect of ZOL appears to be dependent on the inhibition of prenylation of small-G-proteins. From these findings, we conclude that ZOL could be a potential anticancer agent by inhibiting angiogenesis.


Subject(s)
Bone Density Conservation Agents/pharmacology , Cell Differentiation/drug effects , Diphosphonates/pharmacology , Endothelium, Vascular/cytology , Imidazoles/pharmacology , Mesenchymal Stem Cells/cytology , Neovascularization, Physiologic/drug effects , Antigens, CD/metabolism , Apoptosis/drug effects , Cadherins/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Endothelium, Vascular/drug effects , Endothelium, Vascular/metabolism , Humans , Mesenchymal Stem Cells/drug effects , Mesenchymal Stem Cells/metabolism , Vascular Endothelial Growth Factor Receptor-2/metabolism , Zoledronic Acid
6.
Hepatogastroenterology ; 55(84): 930-5, 2008.
Article in English | MEDLINE | ID: mdl-18705300

ABSTRACT

BACKGROUND/AIMS: The p38 mitogen-activated protein kinases (p38 MAPKs) function in a wide variety of signaling pathways. However, the role of p38s is cell type- and stimulus-dependent. The present study aimed to evaluate the effects of p38 MAPK inhibitor on human colon cancer cells. METHODOLOGY: The effect of p38 MAPK inhibitor, FR167653, on DLD-1 and SW480 was investigated related to cell proliferation, apoptosis induction and caspase activity. Additionally, the effect of FR167653 on colon cancer cell migration, MMPs production and ability to adhere to extracellular matrix was investigated. RESULTS: Inhibitor of p38 MAPK dose-dependently suppressed the proliferative activity of both cell lines, and increased the induction of cell apoptosis. The caspase-3, 8, and 9 activities were accompanied in the pathway. Neither cell migration, MMPs production, nor the ability to adhere extracellular matrix were affected by FR167653. CONCLUSIONS: Inhibitor of p38 MAPK suppressed the proliferation of colon cancer cells by induction of cell apoptosis through the caspase activation. The present results suggest the pro-oncogenic role ofp38 in colon cancer, and its inhibition would be a novel strategy for the prevention and treatment of colon cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Colonic Neoplasms/pathology , Pyrazoles/pharmacology , Pyridines/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , Caspases/metabolism , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Humans , Tumor Cells, Cultured/drug effects , p38 Mitogen-Activated Protein Kinases/metabolism
7.
J Allergy Clin Immunol ; 121(1): 209-14, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17935769

ABSTRACT

BACKGROUND: Epigallocatechin gallate (EGCG), a component of green tea catechin with the strongest biological activity, has been focused in recent years because of its anti-inflammatory and immunomodulatory activities. Dendritic cells (DCs) are professional antigen-presenting cells, capable of priming naive T cells, and play the key roles in the activation of T-cell-mediated immune responses. OBJECTIVE: We aimed to investigate the effect of EGCG on human monocyte-derived DCs (MODCs) and, consequently, on the T-cell-mediated immune response. METHODS: The induction of apoptosis, and the detailed phenotypic and functional changes of MODCs, generated by culture of peripheral blood monocytes in the presence of GM-CSF and IL-4, induced by EGCG was investigated and compared with the effects of dexamethasone. RESULTS: Epigallocatechin gallate induced apoptosis and affected the phenotype of the developing DCs. The expressions of CD83, CD80, CD11c, and MHC class II, which are molecules essential for antigen presentation by DCs, were downregulated by EGCG. EGCG also suppressed the endocytotic ability of immature DCs, whereas dexamethasone-treated DCs had higher endocytotic ability than control DCs. Most importantly, mature DCs treated with EGCG inhibited stimulatory activity toward allogeneic T cells while secreting high amounts of IL-10. CONCLUSION: Epigallocatechin gallate induces immunosuppressive alterations on human MODCs, both by induction of apoptosis and suppression of cell surface molecules and antigen presentation.


Subject(s)
Antigen-Presenting Cells/drug effects , Catechin/analogs & derivatives , Cell Differentiation/drug effects , Dendritic Cells/drug effects , Immunosuppression Therapy , Apoptosis/drug effects , Catechin/pharmacology , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/physiology , Humans , Interleukin-10/metabolism , Monocytes/cytology , T-Lymphocytes/immunology
8.
Planta Med ; 73(10): 1068-73, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17691055

ABSTRACT

Although some isoprenoids, such as taxans and geranylgeraniol (GGOH), have been reported to have strong anticancer activities, the effect of plaunotol, the isoprenoid extracted from the leaves of Plau-noi, on cancer has not yet been evaluated. Here, we aimed to investigate the effect of plaunotol on gastric cancer cell lines. Three gastric cancer cell lines, namely MKN-45, MKN-74 and AZ-521 were used. Plaunotol was tested at 10, 20, 30 and 40 micromol/L. Plaunotol dose-dependently inhibited the growth of all gastric cancer cells, dependent on the induction of apoptosis. Caspases-8, -9 and -3, were found to be activated in the apoptotic cells. The expression of Bax protein was increased, but Bcl-2 and Bcl-xL protein expressions were not significantly affected. Plaunotol should be a promising new antitumor agent, and since it is already available for clinical use in Japan, its anticancer properties should be confirmed in clinical trials.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis/drug effects , Croton , Fatty Alcohols/pharmacology , Phytotherapy , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/therapeutic use , Caspases/metabolism , Cell Line, Tumor/drug effects , Diterpenes , Dose-Response Relationship, Drug , Fatty Alcohols/administration & dosage , Fatty Alcohols/therapeutic use , Flow Cytometry , Humans , In Situ Nick-End Labeling , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Stomach Neoplasms/pathology
9.
Angiogenesis ; 9(2): 83-91, 2006.
Article in English | MEDLINE | ID: mdl-16821112

ABSTRACT

Sulforaphane (SUL), one of the isothiocyanates (ITCs), has recently been focused due to its inhibitory effects on tumor cell growth in vitro and in vivo, which is dependent on the direct effect on cancer cells. In the present study, we aimed to investigate the potential anti-angiogenic effect of SUL and its mechanism of action. Using the human umbilical vein endothelial cells (HUVECs) as a model of angiogenesis, we investigated the effect of SUL on the various steps of angiogenesis, including the proliferation of endothelial cells, tubular formation, and matrix metalloproteinase (MMP) production. Sulforaphane induced a dose-dependent decrease in the proliferative activity of endothelial cells, which was dependent on cell apoptosis. Also SUL inhibited tube formation on matrigel, but did not affect MMP production. The present results demonstrate the anti-angiogenic activity of SUL and its potential use as an anti-cancer drug is suggested.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Apoptosis/drug effects , Cell Proliferation/drug effects , Endothelium, Vascular/drug effects , Thiocyanates/pharmacology , Cell Culture Techniques , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Isothiocyanates , Sulfoxides , Time Factors , Umbilical Veins/cytology
10.
Eur J Cancer ; 42(5): 668-73, 2006 Mar.
Article in English | MEDLINE | ID: mdl-16427272

ABSTRACT

Id genes (inhibitor of DNA binding/differentiation) play important roles in tumour growth. We have previously described crucial roles of Id gene over-expression in endothelial cells for tumour angiogenesis. Here, we have evaluated direct effects of Id gene down-regulation on tumour cells, namely on cell proliferation, motility, and adhesion to lung microvasculature during haematogenous metastasis. For this purpose, Id genes were stably down-regulated by RNA interference in human colorectal cancer cells. These cells showed delayed proliferation, inhibited motility and decreased expression of integrin alpha6 and consequently reduced adhesion to lung microvasculature in mice. Static adhesion assays and laminar flow assays revealed decreased laminin binding capacity of these cells, and blocking experiments confirmed that it could be attributed to decreased expression of integrin alpha6. The present results indicate important roles of Id genes in tumour cells during early steps of haematogenous metastasis and suggest dual effects from their therapeutic inhibition.


Subject(s)
Colorectal Neoplasms/genetics , Hematologic Neoplasms/etiology , Inhibitor of Differentiation Proteins/genetics , Animals , Cell Adhesion Molecules , Cell Line, Tumor , Cell Transformation, Neoplastic , Down-Regulation , Female , Flow Cytometry , Humans , Laminin/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude
11.
Cancer Immunol Immunother ; 55(6): 708-16, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16133110

ABSTRACT

Endothelial progenitor cells (EPCs) have been recently found to exist circulating in peripheral blood of adults, and home to sites of neovascularization in peripheral tissues. They can also be differentiated from peripheral blood mononuclear cells (PBMNCs). In tumor tissues, EPCs are found in highly vascularized lesions. Few reports exist in the literature concerning the characteristics of EPCs, especially related to their surface antigen expressions, except for endothelial markers. Here, we aimed to investigate the surface expression of differentiation markers, and the functional activities of early-outgrowth of EPCs (EO-EPCs), especially focusing on their antigen-presenting ability. EO-EPCs were generated from PBMNCs, by culture in the presence of angiogenic factors. These EO-EPCs had the morphological and functional features of endothelial cells and, additionally, they shared antigen-presenting ability. They induced the proliferation of allogeneic lymphocytes in a mixed-lymphocyte reaction, and could generate cytotoxic lymphocytes, with the ability to lyze tumor cells in an antigen-specific manner. The antigen-presenting ability of EO-EPCs, however, was weaker than that of monocyte-derived dendritic cells, but stronger than peripheral blood monocytes. Since EO-EPCs play an important role in the development of tumor angiogenesis, targeting EPCs would be an effective anti-angiogenic strategy. Alternatively, due to their antigen-presenting ability, EO-EPCs can be used as the effectors of anti-tumor immunotherapy. Since they share endothelial antigens, the activation of a cellular immunity against angiogenic vessels can be expected. In conclusion, EO-EPCs should be an interesting alternative for the development of new therapeutic strategies to combat cancer, either as the effectors or as the targets of cancer immunotherapy.


Subject(s)
Antigen-Presenting Cells/immunology , Endothelial Cells/immunology , Neovascularization, Pathologic/immunology , Stem Cells/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/metabolism , Antigens, Differentiation , Biomarkers/analysis , Endothelial Cells/cytology , Endothelial Cells/metabolism , Flow Cytometry , Humans , Lymphocyte Activation/immunology , Lymphocyte Culture Test, Mixed , Stem Cells/cytology , Stem Cells/metabolism , T-Lymphocytes/immunology
12.
Gan To Kagaku Ryoho ; 32(11): 1576-7, 2005 Oct.
Article in Japanese | MEDLINE | ID: mdl-16315874

ABSTRACT

Cimetidine, one of the most popular histamine-2 receptor antagonists, has been reported to improve survival in gastrointestinal cancer patients and to activate cell-mediated immune response in surgical patients. NKT cells are a population of T cells that share characteristics with natural killer cells, and their main functions are production of immunoregulatory cytokines and cytolytic activities. In this study, we aimed to investigate the effect of cimetidine on the cell-mediated immunoresponse. Six healthy adult volunteers were given 800 mg of cimetidine per day orally, and their blood samples were taken prior to and at days 1, 3, 5, and 7 days post-administration of cimetidine. Leukocyte counts and differentials were obtained by the conventional hemogram, and the leukocyte subsets were analyzed by flow cytometry. Cimetidine administration caused leukocytosis, dependent on the increase of neutrophils, as well as of the CD3-positive T lymphocytes, and the subset of CD4-positive cells among them. On the other hand, the NK cell subpopulation was decreased, and the NKT cell subpopulation was not affected. The present results suggest that cimetidine is a modulator of the cellular immunity, and may be used as the activator of the tumor specific immunoresponse.


Subject(s)
CD4 Lymphocyte Count , Cimetidine/pharmacology , Histamine H2 Antagonists/pharmacology , Immunologic Factors/pharmacology , Administration, Oral , Cimetidine/administration & dosage , Histamine H2 Antagonists/administration & dosage , Humans , Immunologic Factors/administration & dosage , Killer Cells, Natural/drug effects
13.
Cancer Sci ; 96(11): 784-90, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16271072

ABSTRACT

Inhibitor of DNA binding (Id) proteins are essential for cell differentiation, proliferation, migration, invasion and angiogenesis. Recently, they have been shown to correlate with less differentiated phenotypes, high malignant potential and poor clinical outcome in various kinds of tumors. In an attempt to develop new strategies for the treatment of peritoneal metastasis of gastric cancer, we prepared an Id1, 3 double-knockdown gastric cancer cell line, MKN45, by RNA interference and investigated its effects on the development of metastatic nodules in the peritoneal cavity. Both cell proliferation and migration capabilities were decreased in Id1, 3 double-knockdown cells, as was their ability to bind to laminin, which could be explained by the decreased expression of integrin alpha6. These are important steps in the metastatic process. In a mouse model, the number of peritoneal metastatic nodules formed by Id1, 3 double-knockdown cells was reduced compared to mock-transfected control cells, as was the size of individual tumors. In this study, we clearly demonstrated that Id1, 3 double-knockdown significantly impaired the ability of gastric cancer cells to form peritoneal metastasis. Id should be considered an ideal target for the treatment and prevention of gastric cancer, and RNA interference is an attractive and promising strategy to achieve it.


Subject(s)
Inhibitor of Differentiation Protein 1/genetics , Inhibitor of Differentiation Protein 1/physiology , Inhibitor of Differentiation Proteins/genetics , Inhibitor of Differentiation Proteins/physiology , Neoplasm Proteins/genetics , Neoplasm Proteins/physiology , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology , Animals , Cell Movement , Cell Proliferation , Disease Models, Animal , Humans , Mice , Neoplasm Metastasis/genetics , Neoplasm Metastasis/physiopathology , RNA Interference , Tumor Cells, Cultured
14.
World J Surg ; 29(11): 1415-20; discussion 1421, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16240061

ABSTRACT

To determine the clinical efficacy of Roux-en-Y reconstruction (RY) after distal gastrectomy, we compared postoperative outcomes of patients who underwent RY or conventional Billroth I reconstruction (B-I). A total of 50 patients were prospectively randomized to either B-I or RY reconstruction, and complications, postoperative course, and nutritional status were compared. Bile reflux and inflammation in the remnant stomach and lower esophagus were evaluated by postoperative follow-up endoscopy at 6 months. Operative time and blood loss as well as postoperative nutrition did not show significant differences between the two groups. As anticipated, 5 of 24 patients with RY reconstruction developed gastrojejunal stasis in the early postoperative period, which led to a longer postoperative hospital stay as compared with the B-I group (mean +/- S.D; B-I; 19.0 +/- 6.2, RY; 31.8 +/- 21.7 days) (P < 0.05). Endoscopic examination revealed that the frequency of bile reflux (P < 0.01) and degree of inflammation in the remnant stomach (P < 0.05) were less in the RY group than in the B-I group. However, inflammatory findings in the lower esophagus were observed in 7 (27%) of B-I, and 8 (35%) of the RY group, suggesting that late phase esophagitis was not improved in the RY group. Roux-en-Y reconstruction was effective in preventing duodenogastric reflux and resulting gastritis, but it did not prevent esophagitis. Because RY reconstruction induces the frequent complication of Roux-en-Y stasis, causing longer postoperative hospital stay, this method has limited advantages over B-I anastomosis after distal gastrectomy.


Subject(s)
Gastrectomy/methods , Stomach Neoplasms/surgery , Anastomosis, Surgical/methods , Blood Loss, Surgical , Humans , Length of Stay , Nutritional Status , Prospective Studies , Treatment Outcome
16.
Anticancer Drugs ; 16(4): 401-7, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15746576

ABSTRACT

We have investigated a potential anti-angiogenic effect of plaunotol, an extract from the leaves of Plau-noi, in an angiogenesis model consisting of human umbilical vein endothelial cells (HUVECs). Plaunotol inhibited the proliferative activity of HUVECs in a dose-dependent manner. In addition, it caused a remarkable decrease of the ability of HUVECs to adhere and spread on gelatin and vitronectin, but not fibronectin. Tube-like formation in Matrigel was also inhibited in a dose-dependent way. These results strongly suggest the specific inhibition of integrin alphavbeta3 to be the main event of plaunotol-induced suppression of angiogenesis. The alphavbeta3 antagonists are known to be potent inhibitors of tumor angiogenesis and plaunotol, by causing the functional inhibition of alphavbeta3, should be considered a promising new anti-angiogenic drug.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cell Proliferation/drug effects , Endothelial Cells/drug effects , Fatty Alcohols/pharmacology , Integrin alphaVbeta3/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Collagen/metabolism , Diterpenes , Dose-Response Relationship, Drug , Drug Combinations , Fibronectins/metabolism , Gelatin/metabolism , Humans , Laminin/metabolism , Proteoglycans/metabolism , Umbilical Veins/cytology , Vitronectin/metabolism
17.
Cancer Sci ; 96(2): 93-9, 2005 Feb.
Article in English | MEDLINE | ID: mdl-15723653

ABSTRACT

High level expression of cyclooxygenase (COX)-2 is reported in 80-90% of colorectal adenocarcinomas. In the recent years, selective inhibitors of COX-2 have been developed, and are shown to effectively protect against cancer development and progression. Colon cancer cells, as well as the epithelial cells in general, are dependent on appropriate interactions with the extracellular matrix (ECM) proteins to achieve a number of important functions, such as proliferation, differentiation, invasion and survival. These interactions are mediated via a family of cell-surface receptors called integrins, which interact with cytoskeletal proteins on the cytoplasmic side of the plasma membrane and thereby provide a link between the ECM and the cytoskeleton. In the present study, a high-COX-2 (high level COX-2 expression) colon cancer cell line, HT-29, and a low-COX-2 (low level COX-2 expression), DLD-1, were used to investigate the anticolon cancer effect of the selective COX-2 inhibitor, JTE-522. Moreover, to clarify its mechanisms of action, we focused especially on the ability to adhere to and to migrate on ECM. We could clearly demonstrate that, in addition to the decrease of the proliferative activity, JTE-522 caused a dose-dependent decrease in both the ability of colon cancer cells to adhere to and to migrate on ECM. These effects were, at least in part, dependent on the down-regulation of beta1-integrin expression, which was evident in HT-29, the high-COX-2 colon cancer cells, but not the low-COX-2, DLD-1. In addition, prostaglandin E2 almost completely reversed the effect of JTE-522, strongly suggesting the involvement of a COX-2-dependent pathway. In conclusion, for the first time, we could demonstrate the down-regulation of beta1 integrin caused by COX-2 inhibition, with consequent impairment of the ability of cancer cells to adhere to and to migrate on ECM, which are crucial steps for cancer metastases to develop.


Subject(s)
Benzenesulfonates/pharmacology , Integrin beta1/metabolism , Oxazoles/pharmacology , Benzenesulfonates/antagonists & inhibitors , Cell Adhesion/drug effects , Cell Division/drug effects , Cell Movement/drug effects , Colonic Neoplasms , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/pharmacology , Dinoprostone/pharmacology , Dose-Response Relationship, Drug , Down-Regulation , Extracellular Matrix/drug effects , HT29 Cells , Humans , Membrane Proteins , Oxazoles/antagonists & inhibitors , Prostaglandin-Endoperoxide Synthases , Tumor Cells, Cultured
18.
J Allergy Clin Immunol ; 115(1): 186-91, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15637567

ABSTRACT

BACKGROUND: Monocytes are the main effector cells of the immune system, and the regulation of their survival and apoptosis is essential for monocyte-involved immune responses. Green tea polyphenol catechin has been reported to have antiallergic and anti-inflammatory activities, but its effect on monocytes has not yet been explored. OBJECTIVE: To elucidate the mechanisms of the anti-inflammatory effect of catechin, we studied the effect of catechin, especially epigallocatechin gallate (EGCG), on the apoptosis of monocytes. METHODS: Isolated peripheral blood monocytes were incubated without or with catechin, and apoptosis was evaluated by annexin V and propidium iodide double-staining or terminal deoxynucleotidyl assay. The activation of caspases 3, 8, and 9 was also evaluated by flow cytometry. The influence of GM-CSF or LPS, the known monocyte survival factors, on the EGCG-induced apoptosis of monocytes was investigated. RESULTS: Among the 4 catechin derivatives tested, EGCG and epicatechin gallate induced apoptosis of monocytes. Caspases 3, 8, and 9, which play a central role in the apoptotic cascade, were dose-dependently activated by EGCG treatment. The EGCG-induced apoptosis of monocytes was not affected by GM-CSF or LPS. CONCLUSION: Catechin, especially EGCG, by promoting monocytic apoptosis, may be a new promising anti-inflammatory agent, and should be tested in clinical trials.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Apoptosis/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Monocytes/drug effects , Caspases/metabolism , Cells, Cultured , Dose-Response Relationship, Drug , Humans , Monocytes/immunology , Monocytes/physiology , Tea
19.
Int J Cancer ; 113(4): 541-8, 2005 Feb 10.
Article in English | MEDLINE | ID: mdl-15472909

ABSTRACT

High-level expression of cyclooxygenase (COX)-2 is reported in 80-90% of colorectal adenocarcinomas. Selective inhibition of COX-2 was shown to reduce colorectal tumorigenesis in different models of carcinogenesis and to prevent metastasis in xenograft tumor models, as well as to suppress in vitro induced angiogenesis. Recently, COX-2 was reported to be expressed not only in malignant epithelial cells, but also in the neovasculature that feeds the tumor in a variety of solid human cancers. Thus, one of the possible mechanisms by which selective COX-2 inhibitor reduces tumor growth and metastasis is through inhibition of tumor angiogenesis. Although a report suggested a possible role of endothelial COX-1 in the process of angiogenesis, in a recent study, the selective inhibition of COX-2 was shown to strongly inhibit angiogenesis by inducing endothelial cell (EC) apoptosis. In the present study, using human umbilical vein endothelial cells (HUVECs) as a model of angiogenesis, we investigated the potential antiangiogenic effect of the selective COX-2 inhibitor and its mechanism of action, and clearly demonstrated that selective inhibition of COX-2 caused a dose-dependent decrease in the proliferative activity of ECs, as well as an inhibition of capillary-like tube formation. The inhibitory effect on EC proliferation was dependent on the cell cycle arrest to the G1 phase and not on cell apoptosis.


Subject(s)
Cyclooxygenase Inhibitors/pharmacology , Endothelium, Vascular/pathology , G1 Phase/drug effects , Isoenzymes/antagonists & inhibitors , Apoptosis/drug effects , Benzenesulfonates/pharmacology , Cell Adhesion , Cell Proliferation , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Dose-Response Relationship, Drug , Extracellular Matrix/metabolism , Humans , Matrix Metalloproteinase 2/metabolism , Membrane Proteins , Neovascularization, Pathologic/prevention & control , Oxazoles/pharmacology , Prostaglandin-Endoperoxide Synthases , Umbilical Veins
20.
J Allergy Clin Immunol ; 113(6): 1211-7, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15208607

ABSTRACT

BACKGROUND: Although green tea polyphenol catechin has been reported to have antiallergic and anti-inflammatory activities, the precise mechanisms of its effect on the immune system have been poorly investigated. OBJECTIVE: In this study, we aimed to elucidate the mechanisms of the anti-inflammatory effect of catechin. For this purpose, we studied the effect of 2 kinds of catechin, epigallocatechin gallate (EGCG) and epicatechin gallate, on peripheral blood CD8+ T cells, which play the key role in immune responses. METHODS: Isolated peripheral blood mononuclear cells or CD8+ T cells were incubated without or with catechin, and the changes in the surface expression of integrin molecules were investigated by flow cytometry and the direct binding of catechin to CD11b molecule by competitive ELISA. Also, the effect of catechin on the ability of CD8+ T cells to bind intracellular adhesion molecule 1 and to migrate in response to chemokines was evaluated by using the adhesion and migration assays. RESULTS: The 2 catechins directly bound to CD11b expressed on CD8+ T cells, which caused a consequent decrease of flow-cytometric CD11b expression. The effect was more prominent with EGCG than epicatechin gallate, and the impaired expression of CD11b induced by EGCG resulted in decreased ability of CD8+ T cells to adhere intercellular adhesion molecule 1, and consequently decreased migration in response to chemokines. CONCLUSION: We concluded that catechin, especially EGCG, by downregulating CD11b expression on CD8+ T cells and, in consequence, inhibiting infiltration of these cells into the sites of inflammation, is a promising new potent anti-inflammatory agent.


Subject(s)
Antigens, CD1/metabolism , CD8-Positive T-Lymphocytes/drug effects , Catechin/analogs & derivatives , Catechin/pharmacology , Antigens, CD1/analysis , CD8-Positive T-Lymphocytes/physiology , Catechin/metabolism , Cell Adhesion/drug effects , Cell Movement/drug effects , Cells, Cultured , Enzyme-Linked Immunosorbent Assay , Granulocytes/chemistry , Granulocytes/drug effects , Humans , Integrins/analysis , Monocytes/chemistry , Monocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL
...