Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 20
Filtrer
Plus de filtres











Base de données
Gamme d'année
1.
Radiother Oncol ; 200: 110480, 2024 Aug 17.
Article de Anglais | MEDLINE | ID: mdl-39159681

RÉSUMÉ

BACKGROUND AND PURPOSE: Local radiotherapy (RT) exerts immunostimulatory effects by inducing immunogenic cell death. However, it remains unknown whether in vitro-irradiated tumor cells can elicit anti-tumor responses and enhance the efficacy of local RT and immune checkpoint inhibitors when injected in vivo. METHODS AND MATERIALS: We tested the "in vitro-irradiated cancer vaccine (ICV)", wherein tumor cells killed by varying doses of irradiation and their supernatants are intravenously injected. We examined the efficacy of combining local RT (24 Gy in three fractions), PD-L1 blockade, and the ICV in a murine breast cancer model. The immune cell profiles were analyzed via flow cytometry and immunohistochemistry. The cytokine levels were measured by multiplex immunoassays. RESULTS: The ICV significantly increased the effector memory phenotype and interferon-γ production capacity in splenic CD8+ T cells. The in vitro-irradiated products contained immune response-related molecules. When combined with local RT and PD-L1 blockade, the ICV significantly delayed the growth of irradiated and non-irradiated tumors. The triple combination therapy increased the proportions of CD8+ T cells and effector memory CD8+ T cells while decreasing the proportion of CTLA-4+ exhausted CD8+ T cells within tumor microenvironment. Additionally, plasma level of interferon-γ and proliferation of effector T cells in the spleen and tumor-draining lymph nodes were significantly increased by the triple combination therapy. CONCLUSIONS: The ICV enhanced the therapeutic efficacy of local RT and PD-L1 blockade by augmenting anti-tumor immune responses. Our findings suggest a therapeutic potential of in vitro-irradiation products of tumor cells.

2.
Radiother Oncol ; 190: 109981, 2024 Jan.
Article de Anglais | MEDLINE | ID: mdl-37925106

RÉSUMÉ

PURPOSE: In this study, we investigated whether local radiotherapy (RT) and an anti-glucocorticoid-induced tumor necrosis factor receptor (GITR) agonist could increase the efficacy of PD-L1 blockade. METHODS AND MATERIALS: We analyzed a breast cancer dataset from the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) to determine the role of GITR in breast cancer. We used the 4T1 murine TNBC model (primary and secondary tumors) to investigate the efficacy of PD-L1 blockade, local RT, anti-GITR agonist, and their combinations. We assessed tumor growth by tumor volume measurements, in vivo bioluminescence imaging, and metastatic lung nodule counts to evaluate the effects of these treatments. Flow cytometry and immunohistochemistry determined the proportions and phenotypes of CD8+ T-cells and regulatory T-cells (Tregs) in the tumors and spleen. Plasma cytokine levels were measured by enzyme-linked immunosorbent assay. RESULTS: In the METABRIC cohort, patients with high expression of TNFRSF18, which encodes GITR, had significantly better survival than those with low expression. Adding local RT or anti-GITR agonist to PD-L1 blockade did not significantly augment efficacy compared to PD-L1 blockade alone; however, adding both to PD-L1 blockade significantly reduced tumor growth and lung metastasis. The benefits of the triple combination were accompanied by increased CD8+ T-cells and decreased Tregs in the tumor microenvironment and spleen. CONCLUSIONS: The combination of local RT and an anti-GITR agonist significantly enhanced the anti-tumor immune responses induced by PD-L1 blockade. These results provide the preclinical rationale for the combination of therapy.


Sujet(s)
Antigène CD274 , Tumeurs du sein , Humains , Animaux , Souris , Femelle , Lymphocytes T CD8+ , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/radiothérapie , Glucocorticoïdes/pharmacologie , Récepteurs aux facteurs de nécrose tumorale , Microenvironnement tumoral , Lignée cellulaire tumorale
3.
Int J Mol Sci ; 24(17)2023 Aug 22.
Article de Anglais | MEDLINE | ID: mdl-37685868

RÉSUMÉ

Bromodomain-containing protein 4 (BRD4) is an intracellular protein that regulates expression of various cellular functions. This study investigated whether BRD4 inhibition can alter the immunomodulatory and antitumor effects of radiation therapy (RT). A murine breast cancer cell line was implanted into BALB/c mice. The dual-tumor model was used to evaluate the abscopal effects of RT. A total of 24 Gy was delivered and BRD4 inhibitor was injected intravenously. Tumor size was measured, and in vivo imaging was performed to evaluate tumor growth. Flow cytometry and immunohistochemistry were performed to examine immunologic changes upon treatment. The combination of BRD4 inhibitor and RT significantly suppressed tumor growth compared to RT alone. BRD4 inhibitor reduced the size of the unirradiated tumor, indicating that it may induce systemic immune responses. The expression of HIF-1α and PD-L1 in the tumor was significantly downregulated by the BRD4 inhibitor. The proportion of M1 tumor-associated macrophages (TAMs) increased, and the proportion of M2 TAMs decreased upon BRD4 inhibition. BRD4 inhibitor expanded CD4+ and CD8+ T cell populations in the tumor microenvironment. Additionally, splenic monocytic myeloid derived suppressor cells, which were increased by RT, were reduced upon the addition of BRD4 inhibitor. Therefore, the addition of BRD4 inhibitor significantly enhanced the systemic antitumor responses of local RT.


Sujet(s)
Tumeurs du sein , Tumeurs , Protéines nucléaires , Animaux , Souris , Lymphocytes T CD8+ , Cytométrie en flux , Immunomodulation , Souris de lignée BALB C , Modèles animaux de maladie humaine , Tumeurs du sein/traitement médicamenteux , Tumeurs du sein/radiothérapie
4.
J Korean Soc Radiol ; 83(5): 1104-1108, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-36276207

RÉSUMÉ

Myocardial calcification can develop owing to several conditions. It is a rare complication following sepsis and renal failure. We report a case of rapid development of left ventricular mid-wall calcification observed using CT and cardiac MRI in a patient after recovery from sepsis and acute renal failure.

5.
Cancers (Basel) ; 14(11)2022 May 29.
Article de Anglais | MEDLINE | ID: mdl-35681672

RÉSUMÉ

Immune checkpoint inhibitors have been successful in a wide range of tumor types but still have limited efficacy in immunologically cold tumors, such as breast cancers. We hypothesized that the combination of agonistic anti-OX40 (α-OX40) co-stimulation, PD-1 blockade, and radiotherapy would improve the therapeutic efficacy of the immune checkpoint blockade in a syngeneic murine triple-negative breast cancer model. Murine triple-negative breast cancer cells (4T1) were grown in immune-competent BALB/c mice, and tumors were irradiated with 24 Gy in three fractions. PD-1 blockade and α-OX40 were administered five times every other day. Flow cytometric analyses and immunohistochemistry were used to monitor subsequent changes in the immune cell repertoire. The combination of α-OX40, radiotherapy, and PD-1 blockade significantly improved primary tumor control, abscopal effects, and long-term survival beyond 2 months (60%). In the tumor microenvironment, the ratio of CD8+ T cells to CD4 + FOXP3+ regulatory T cells was significantly elevated and exhausted CD8+ T cells (PD-1+, CTLA-4+, TIM-3+, or LAG-3+ cells) were significantly reduced in the triple combination group. Systemically, α-OX40 co-stimulation and radiation significantly increased the CD103+ dendritic cell response in the spleen and plasma IFN-γ, respectively. Together, our results suggest that the combination of α-OX40 co-stimulation and radiation is a viable approach to overcome therapeutic resistance to PD-1 blockade in immunologically cold tumors, such as triple-negative breast cancer.

6.
Eur J Cancer ; 157: 450-463, 2021 11.
Article de Anglais | MEDLINE | ID: mdl-34601286

RÉSUMÉ

INTRODUCTION: We hypothesised that the combined use of radiation therapy and a phosphoinositide 3-kinaseγδ inhibitor to reduce immune suppression would enhance the efficacy of an immune checkpoint inhibitor. METHODS: Murine breast cancer cells (4T1) were grown in both immune-competent and -deficient BALB/c mice, and tumours were irradiated by 3 fractions of 24 Gy. A PD-1 blockade and a phosphoinositide 3-kinase (PI3K)γδ inhibitor were then administered every other day for 2 weeks. The same experiments were performed in humanised patient-derived breast cancer xenograft model and its tumour was sequenced to identify immune-related pathways and profile infiltrated immune cells. Transcriptomic and clinical data were acquired from The Cancer Genome Atlas pan-cancer cohort, and the deconvolution algorithm was used to profile immune cell repertoire. RESULTS: Using a PI3Kγδ inhibitor, radiation therapy (RT) and PD-1 blockade significantly delayed primary tumour growth, boosted the abscopal effect and improved animal survival. RT significantly increased CD8+cytotoxic T-cell fractions, immune-suppressive regulatory T cells (Tregs), myeloid-derived suppressor cells and M2 tumour-associated macrophages (TAMs). However, the PI3Kγδ inhibitor significantly lowered the proportions of Tregs, myeloid-derived suppressor cells and M2 TAMs, achieving dramatic gains in splenic, nodal, and tumour CD8+ T-cell populations after triple combination therapy. In a humanised patient-derived breast cancer xenograft model, triple combination therapy significantly delayed tumour growth and decreased immune-suppressive pathways. In The Cancer Genome Atlas cohort, high Treg/CD8+ T cell and M2/M1 TAM ratios were associated with poor overall patient survival. CONCLUSION: These findings indicate PI3Kγ and PI3Kδ are clinically relevant targets in an immunosuppressive TME, and combining PI3Kγδ inhibitor, RT and PD-1 blockade may overcome the therapeutic resistance of immunologically cold tumours. SYNOPSIS: Combining PI3Kγδ inhibitor, RT, and PD-1 blockade may be a viable clinical approach, helping to overcome the therapeutic resistance of immunologically cold tumours such as breast cancer.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/pharmacologie , Tumeurs du sein/thérapie , Chimioradiothérapie/méthodes , Inhibiteurs de points de contrôle immunitaires/pharmacologie , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Animaux , Protocoles de polychimiothérapie antinéoplasique/usage thérapeutique , Tumeurs du sein/génétique , Tumeurs du sein/immunologie , Tumeurs du sein/anatomopathologie , Lignée cellulaire tumorale , Phosphatidylinositol 3-kinases de classe I/métabolisme , Phosphatidylinositol 3-kinases de classe Ib/métabolisme , Modèles animaux de maladie humaine , Résistance aux médicaments antinéoplasiques/effets des médicaments et des substances chimiques , Résistance aux médicaments antinéoplasiques/immunologie , Résistance aux médicaments antinéoplasiques/effets des radiations , Synergie des médicaments , Femelle , Humains , Inhibiteurs de points de contrôle immunitaires/usage thérapeutique , Souris , Inhibiteurs des phosphoinositide-3 kinases/usage thérapeutique , Récepteur-1 de mort cellulaire programmée/antagonistes et inhibiteurs , Lymphocytes T cytotoxiques/effets des médicaments et des substances chimiques , Lymphocytes T cytotoxiques/immunologie , Lymphocytes T cytotoxiques/effets des radiations , Échappement de la tumeur à la surveillance immunitaire/effets des médicaments et des substances chimiques , Échappement de la tumeur à la surveillance immunitaire/effets des radiations , Microenvironnement tumoral/effets des médicaments et des substances chimiques , Microenvironnement tumoral/immunologie , Microenvironnement tumoral/effets des radiations , Tests d'activité antitumorale sur modèle de xénogreffe
7.
Int J Radiat Oncol Biol Phys ; 110(3): 845-858, 2021 07 01.
Article de Anglais | MEDLINE | ID: mdl-33642128

RÉSUMÉ

PURPOSE: The poor response of breast cancer to immune checkpoint blockade might result from low immunogenicity and the immune-suppressive tumor microenvironment. We hypothesized that in situ tumor vaccination via radiation therapy (RT) and suppression of immune tolerance via phosphoinositide 3-kinase δ (PI3Kδ) inhibition would enhance the efficacy of immune checkpoint blockade. METHODS AND MATERIALS: 4T1 murine breast cancer cells were grown in both immune-competent and -deficient BALB/c mice, and tumors were irradiated with 24 Gy in 3 fractions. A PD-1 blockade and a PI3Kαδ inhibitor were then administered every other day for 2 weeks. Fluorescence-activated cell sorting and immunohistochemistry served to monitor subsequent changes in immune cell repertoire. RESULTS: The triple combination of RT, PD-1 blockade, and PI3Kαδ inhibitor significantly delayed tumor growth. The immune-deficient syngeneic 4T1 murine tumor model failed to show this tumor growth delay. Use of RT and PI3Kαδ inhibitor increased the proportions of CD8+ T cells; PI3Kαδ inhibitor led to a decrease in regulatory T cells and polymorphonuclear myeloid-derived suppressor cells. The triple combination resulted in a remarkable increase in cytotoxic CD8+ T cells, suggesting a prominent immune-modulatory effect. The abscopal effect was most prominent in the triple-combination therapy group, and it correlated with splenic CD8+ T cell accumulation. CONCLUSIONS: These findings collectively indicate that combining RT, PI3Kαδ inhibitor, and PD-1 blockade could be a viable approach, helping to overcome the therapeutic resistance of immunologically cold tumors, such as breast cancer, with an immunosuppressive tumor microenvironment.


Sujet(s)
Phosphatidylinositol 3-kinases/métabolisme , Inhibiteurs des phosphoinositide-3 kinases/pharmacologie , Récepteur-1 de mort cellulaire programmée/immunologie , Tumeurs du sein triple-négatives/immunologie , Tumeurs du sein triple-négatives/thérapie , Animaux , Lignée cellulaire tumorale , Humains , Souris , Souris de lignée BALB C , Inhibiteurs des phosphoinositide-3 kinases/usage thérapeutique , Tumeurs du sein triple-négatives/traitement médicamenteux , Tumeurs du sein triple-négatives/radiothérapie , Tests d'activité antitumorale sur modèle de xénogreffe
8.
Medicine (Baltimore) ; 98(8): e14536, 2019 Feb.
Article de Anglais | MEDLINE | ID: mdl-30813160

RÉSUMÉ

RATIONALE: Advanced ovarian malignancies are associated with poor overall survival; thus, patients often turn to alternative treatments, despite the controversy surrounding their use. Mistletoe extract has been commonly used as complementary medicine to treat patients with cancer for several decades, and has proven benefits in integrative oncology. PATIENT CONCERNS: A 47-year-old woman with stage IVB ovarian cancer who underwent optimal surgical cytoreduction, but whose disease persisted after adjuvant platinum-based combination chemotherapy and 2nd-line chemotherapy. DIAGNOSIS AND INTERVENTIONS: The patient discontinued chemotherapy due to her septic condition and acute kidney injury accompanied by acute pyelonephritis, and opted for adjuvant treatment with mistletoe extract. OUTCOMES: The patient has achieved good health without progression of cancer or ascites over the 42 months since the 1st diagnosis and 24 months since the last relapse. LESSIONS: Our case suggests that mistletoe extract can produce favorable outcomes in patients with platinum-refractory ovarian cancer.


Sujet(s)
Protocoles de polychimiothérapie antinéoplasique/effets indésirables , Gui/effets des médicaments et des substances chimiques , Récidive tumorale locale/thérapie , Tumeurs de l'ovaire/thérapie , Phytothérapie/méthodes , Protocoles de polychimiothérapie antinéoplasique/administration et posologie , Traitement médicamenteux adjuvant/effets indésirables , Traitement médicamenteux adjuvant/méthodes , Interventions chirurgicales de cytoréduction/effets indésirables , Interventions chirurgicales de cytoréduction/méthodes , Femelle , Humains , Adulte d'âge moyen , Ovaire/anatomopathologie , Composés du platine/effets indésirables , Composés du platine/usage thérapeutique , Tomodensitométrie
9.
Obstet Gynecol Sci ; 62(2): 120-126, 2019 Mar.
Article de Anglais | MEDLINE | ID: mdl-30918880

RÉSUMÉ

OBJECTIVE: Women with pelvic floor disorders and urinary incontinence (UI) are at an increased risk of sexual dysfunction. The purpose of this study was to investigate the effect of surgery for UI on sexual function. METHODS: We retrospectively reviewed the charts of 82 women who underwent mid-urethral transobturator tape (TOT) surgery between March 2010 and December 2014. The Pelvic Floor Distress Inventory-20 (PFDI-20) and the Pelvic Organ Prolapse/Urinary Incontinence Sexual Function Questionnaire-12 (PISQ-12) were administered pre- and postoperatively. RESULTS: We observed a significant increase in the total postoperative PISQ-12 scores compared to the preoperative scores (from 27.1±7.3 to 30.5±6.8, P<0.001). Improved sexual function was confirmed in the physical (13.3±4.5 vs. 15.8±3.5, P<0.001) and partner-related domains (6.7±2.6 vs. 7.4±2.4, P=0.001). Coital incontinence and preoperative urinary distress inventory score were significant factors influencing postoperative sexual function in women undergoing TOT surgery for UI after adjusting for age, body mass index, menopause, and preoperative PISQ-12 score in multivariate regression analysis. CONCLUSION: TOT surgery for UI correction resulted in significant improvement in sexual function.

10.
Toxicol Res ; 34(2): 97-102, 2018 Apr.
Article de Anglais | MEDLINE | ID: mdl-29686771

RÉSUMÉ

The objective of this study was to determine the oral wound healing effects of acai berry water extracts (ABWE) in rat oral mucosa. To estimate the anti-oxidative effects of ABWE, the contents of phenolic compounds, and DPPH (1,1-diphenyl-2-picryl hydrazyl) and ABTS (2,2'-azinobis-(3-ethylbenzothiazoline-6-sulfonic acid)) abilities were evaluated. Wound healing effects of ABWE were tested following 6-day exposure after induction of wound by applying 50% acetic acid to oral mucosa of Sprague-Dawley rats. Macroscopic and histopathological analyses were performed to determine wound healing effects of ABWE. Sodium fusidate (20 mg/g) was used as positive control. ABWE showed significantly high antioxidant effects in all assays, although its potency was weaker than the positive control. From day 3 after treatment, wound healing effects of ABWE were observed in oral mucosa. These wound healing effects were also consistent with histopathological evaluation results. Taken together, these results indicate that ABWE might have potential as an oral wound healing agent in the future.

11.
J Menopausal Med ; 24(3): 163-168, 2018 Dec.
Article de Anglais | MEDLINE | ID: mdl-30671408

RÉSUMÉ

OBJECTIVES: To investigate the rate of postoperative urinary retention (POUR) and identify the risk factors for this complication in women who underwent transvaginal uterosacral suspension surgery. METHODS: A retrospective chart review was conducted for 75 women who underwent transvaginal uterosacral suspension surgery with vaginal hysterectomy, repair of cystocele, and levator myorrhaphy with/without transobturator anti-incontinence surgery. POUR was defined as a need for continuous intermittent catheterization on the third day subsequent to removal of the urethral indwelling catheter. RESULTS: Acute POUR was reported in 18 women (24.0%). Thirty-six of the 75 patients (48.0%) had undergone anti-incontinence surgery. Crude analysis revealed significant association between the following variables and the risk of POUR: hypertension, the lower average flow rate in the pressure-flow study (PFS), greater post-void residual (PVR) urine volume in PFS, and PVR >30% of the total bladder capacity (TBC) in PFS. In the logistic regression analysis, PVR >30% of the TBC in PFS was identified as the only significant predictor of POUR (odds ratio, 15.4; 95% confidence interval, 2.5-90.9; P = 0.003). CONCLUSIONS: The PVR >30% of the TBC in PFS was identified as the only predictive factor of acute POUR in women who underwent transvaginal uterosacral suspension surgery.

12.
Oncotarget ; 8(38): 63901-63910, 2017 Sep 08.
Article de Anglais | MEDLINE | ID: mdl-28969039

RÉSUMÉ

BACKGROUND: Characteristics of tumor microenvironment have been suggested as predictive markers of anti-EGFR or anti-HER2 treatment response. However, the effect of EGFR/HER2 signal blockade on the tumor immune microenvironment is unclear. METHODS: EGFR/HER2 pathway signaling and PD-L1 expression in gastric cancer cell lines were screened by western blot analysis. PD-L1 and HER2 expressions in 251 resected gastric tumors were determined by immunohistochemistry, and changes in EFGR, HER2, and PD-L1 expression in paired specimens between pre- and post-chemotherapy were evaluated. PD-L1 expression in HER2-amplified cell lines was evaluated by western blotting, fluorescence-activated cell sorting, reverse transcription, and real-time quantitative PCR analyses before and after afatinib, lapatinib, pictilisib and trametinib treatment. Changes in cytokines were evaluated by reverse transcription, real-time quantitative PCR, and enzyme-linked immunosorbent assay after EGFR/HER2 inhibition. RESULTS: Cell lines with pEGFR or pHER2 overexpression showed higher PD-L1 expression. In resected gastric tumors, HER2 expression was significantly associated with PD-L1 expression (p=0.030). PD-L1 overexpression accompanied by increased HER2 expression was identified in a post-chemotherapy specimen from a patient with an initial HER2/PD-L1-negative tumor. In HER2-overexpressing cell lines, PD-L1 expression was decreased in a dose- and time-dependent manner after afatinib and lapatinib treatment. PI3K pathway inhibition by pictilisib, but not MEK pathway inhibition by trametinib, resulted in PD-L1 suppression. After lapatinib treatment, the release of CCL2, CCL21, VEGF and CXCL1 decreased in a dose-dependent manner. CONCLUSIONS: Inhibition of the EGFR/HER2 signaling pathway, particularly of downstream PI3K activity, suppressed PD-L1 and release of cytokines, suggesting that EGFR/HER2 inhibition may create a more favorable milieu for tumor immunotherapy.

13.
Toxicol Res ; 33(2): 149-156, 2017 Apr.
Article de Anglais | MEDLINE | ID: mdl-28503264

RÉSUMÉ

The purpose of this study was to investigate the wound healing effect of acai berry water extracts (ABWE) and a possible underlying mechanism involved in its action using various in vitro and in vivo models. The wound healing effect of ABWE was evaluated by migration assay using HS68 fibroblast cells. In addition, its effect on mRNA expression of procollagen, fibronectin, and MMP-1 was determined. Moreover, the wound healing effect of ABWE was evaluated in in vivo wound models through macroscopic and microscopic observation. In addition, mRNA expression levels of wound related genes were determined. Results revealed that ABWE was not cytotoxic. It increased migration of HS68 fibroblast cells. ABWE increased mRNA expression levels of fibronectin but decreased the mRNA expression levels of MMP-1. ABWE also showed significantly potent wound healing effect in vivo based on macroscopic and histopathological observation and mRNA expression evaluation for wound related genes. Taken together, our results indicated that ABWE might have potential as a wound healing agent.

14.
Cancer Res Treat ; 48(1): 355-64, 2016 Jan.
Article de Anglais | MEDLINE | ID: mdl-25761479

RÉSUMÉ

PURPOSE: HM781-36B is a novel and irreversible pan-human epidermal growth factor receptor (HER) inhibitor with TEC cytoplasmic kinase inhibition. The aim of this study is to evaluate the antitumor activity and mechanism of action for HM781-36B in CRC cell lines. MATERIALS AND METHODS: The CRC cell lines were exposed to HM781-36B and/or oxaliplatin (L-OHP), 5-fluorouracil (5-FU), SN-38. The cell viability was examined by Cell Titer-Glo luminescent cell viability assay kit. Change in the cell cycle and protein expression was determined by flow cytometry and immunoblot analysis, respectively. Synergism between 2 drugs was evaluated by the combination index. RESULTS: The addition of HM781-36B induced potent growth inhibition in both DiFi cells with EGFR overexpression and SNU-175 cells (IC50, 0.003 µM and 0.005 µM, respectively). Furthermore, HM781-36B induced G1 arrest of the cell cycle and apoptosis, and reduced the levels of HER family and downstream signaling molecules, pERK and pAKT, as well as nonreceptor/cytoplasmic tyrosine kinase, BMX. The combination of HM781-36B with 5-FU, L-OHP, or SN-38 showed an additive or synergistic effect in most CRC cells. CONCLUSION: These findings suggest the potential roles of HM781-36B as the treatment for EGFR-overexpressing colon cancer, singly or in combination with chemotherapeutic agents. The role of BMX expression as a marker of response to HM781-36B should be further explored.


Sujet(s)
Antinéoplasiques/usage thérapeutique , Tumeurs colorectales/traitement médicamenteux , Tumeurs colorectales/anatomopathologie , Quinazolines/pharmacologie , Quinazolines/usage thérapeutique , Antinéoplasiques/administration et posologie , Apoptose/effets des médicaments et des substances chimiques , Camptothécine/analogues et dérivés , Camptothécine/pharmacologie , Camptothécine/usage thérapeutique , Lignée cellulaire tumorale , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales/métabolisme , Synergie des médicaments , Récepteurs ErbB/antagonistes et inhibiteurs , Extracellular Signal-Regulated MAP Kinases/métabolisme , Fluorouracil/pharmacologie , Fluorouracil/usage thérapeutique , Points de contrôle de la phase G1 du cycle cellulaire/effets des médicaments et des substances chimiques , Humains , Irinotécan , Composés organiques du platine/pharmacologie , Composés organiques du platine/usage thérapeutique , Oxaliplatine , Biosynthèse des protéines/effets des médicaments et des substances chimiques , Protein-tyrosine kinases/antagonistes et inhibiteurs , Protein-tyrosine kinases/métabolisme , Protéines proto-oncogènes c-akt/métabolisme , Quinazolines/administration et posologie
15.
Adv Healthc Mater ; 4(9): 1311-6, 2015 Jun 24.
Article de Anglais | MEDLINE | ID: mdl-25866283

RÉSUMÉ

An enzyme-mimetic nanoparticle-based point of care bioassay device is developed for rapid and sensitive detection of analytes. Digital images acquired by smart cellular phones allow quantifying the amounts of analytes. Using this new device, quantitative analysis of liquid sample is performed within 15 min with an order of magnitude enhancement of sensitivity compared with conventional Au nanoparticle-based devices.


Sujet(s)
Dosage biologique/méthodes , Matériaux biomimétiques/composition chimique , Nanoparticules/composition chimique , Myeloperoxidase/composition chimique , Systèmes automatisés lit malade , Animaux , Dosage biologique/instrumentation , Humains
16.
Cancer Res Treat ; 47(3): 501-8, 2015 Jul.
Article de Anglais | MEDLINE | ID: mdl-25672581

RÉSUMÉ

PURPOSE: p21-activated kinases (PAKs) are involved in cytoskeletal reorganization, gene transcription, cell proliferation and survival, and oncogenic transformation. Therefore, we hypothesized that PAK expression levels could predict the sensitivity of pancreatic cancer cells to gemcitabine treatment, and PAKs could be therapeutic targets. MATERIALS AND METHODS: Cell viability inhibition by gemcitabine was evaluated in human pancreatic cancer cell lines (Capan-1, Capan-2, MIA PaCa-2, PANC-1, Aspc-1, SNU-213, and SNU-410). Protein expression and mRNA of molecules was detected by immunoblot analysis and reverse transcription polymerase chain reaction. To define the function of PAK4, PAK4 was controlled using PAK4 siRNA. RESULTS: Capan-2, PANC-1, and SNU-410 cells were resistant to gemcitabine treatment. Immunoblot analysis of signaling molecules reported to indicate gemcitabine sensitivity showed higher expression of PAK4 and lower expression of human equilibrative nucleoside transporter 1 (hENT1), a well-known predictive marker for gemcitabine activity, in the resistant cell lines. Knockdown of PAK4 using siRNA induced the upregulation of hENT1. In resistant cell lines (Capan-2, PANC-1, and SNU-410), knockdown of PAK4 by siRNA resulted in restoration of sensitivity to gemcitabine. CONCLUSION: PAK4 could be a predictive marker of gemcitabine sensitivity and a potential therapeutic target to increase gemcitabine sensitivity in pancreatic cancer.

17.
Oncol Rep ; 33(1): 185-92, 2015 Jan.
Article de Anglais | MEDLINE | ID: mdl-25370208

RÉSUMÉ

Smad3 and Smad4 are signaling mediators in the transforming growth factor ß (TGFß) pathway and play a major role in the progression and migration of many types of cancers. The TGFß pathway is correlated with resistance against both targeted and conventional chemotherapeutic drugs. The aim of this study was to determine the effect of Smad3/4 on drug sensitivity in chemotherapy-resistant colorectal cancer (CRC) cells. We isolated the TGFß-mediated chemoresistant CRC cell line DLD1-5FU-C10, which showed high expression of Smad3/4 and p21. In order to analyze the influence of Smad3/4 on drug sensitivity in DLD1-5FU-C10 cells, we knocked down Smad3/4 using small interfering RNAs (siRNA). The results showed similar drug sensitivity between the DLD1­5FU-C10 and the DLD1 control cells and reduced p21 expression. In addition, we found a significant increase in the levels of 3 TGFß downstream factors: interleukin 6 (IL6), plasminogen activator (PLAU) and prostaglandin-endoperoxide synthase 2 (PTGS2). Furthermore, we showed that Smad3/4 regulated the JAK1/STAT3 pathway via IL6 in the chemoresistant CRC cell line. In conclusion, we identified Smad3/4 as a novel drug sensitivity regulator in TGFß-mediated chemotherapy-resistant CRC cells. Our results suggest that Smad3/4 regulate p-STAT3 signaling by IL6 and p21 and highlight an important role for STAT3 signaling in Smad3/4 regulated drug sensitivity in chemoresistant CRC cells.


Sujet(s)
Résistance aux médicaments antinéoplasiques , Protéine Smad-3/métabolisme , Protéine Smad-4/métabolisme , Antimétabolites antinéoplasiques/pharmacologie , Lignée cellulaire tumorale , Mouvement cellulaire , Survie cellulaire/effets des médicaments et des substances chimiques , Tumeurs colorectales , Inhibiteur p21 de kinase cycline-dépendante/métabolisme , Fluorouracil/pharmacologie , Régulation de l'expression des gènes tumoraux , Humains , Facteur de transcription STAT-3 , Transduction du signal , Activation de la transcription , Facteur de croissance transformant bêta/physiologie
18.
BMC Cancer ; 14: 241, 2014 Apr 04.
Article de Anglais | MEDLINE | ID: mdl-24708484

RÉSUMÉ

BACKGROUND: Pre-operative chemoradiotherapy (CRT) is the standard treatment in clinical stage T3/4 or node positive rectal cancer. However, there are no established biomarkers that can predict the pathological response and clinical outcome to CRT. METHODS: Immunohistochemical staining was performed in tissue arrays constructed from core tissue specimens taken before treatment and from operative specimens from 112 patients who received 5-FU based pre-operative CRT and surgery. Expression of Ki67, TS, BAX, EpCAM, p53, p21, EGFR, CD44, CD133, CD166, HIF1α and ALDH1 were assessed and correlated with tumor regression grades and disease free survival. RESULTS: Of the 112 patients (M/F 74/38, median age: 62), 20 (17.9%) patients achieved pathologic complete remission (pCR). In analyzing the associations between marker expressions and tumor regression grades, high p21 expression at the pretreatment biopsy was significantly associated with non-pCR (p = 0.022) and poor disease free survival (median DFS - low vs high p21: 75.8 vs 58.1 months, p = 0.002). In the multivariate analysis, high p21 expression level at the pre-treatment biopsy was significantly associated with poor DFS (p = 0.001, HR 6.14; 95% CI 2.03, 18.55). High CD166 expression level at the pretreatment biopsy was also associated with poor DFS (p = 0.003; HR 5.61; 95% CI 1.81, 17.35). CONCLUSION: These show high p21 and CD166 expression at the pretreatment biopsy were associated with tumor regression and poor prognosis in patients treated with 5-FU based CRT. Larger, prospective and functional studies are warranted to determine the role of p21 and CD166 as predictive biomarker of response to CRT.


Sujet(s)
Antigènes CD/biosynthèse , Marqueurs biologiques tumoraux/biosynthèse , Molécules d'adhérence cellulaire neuronale/biosynthèse , Protéines foetales/biosynthèse , Tumeurs du rectum/génétique , Protéines G rho/biosynthèse , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD/génétique , Marqueurs biologiques tumoraux/génétique , Molécules d'adhérence cellulaire neuronale/génétique , Chimioradiothérapie , Survie sans rechute , Femelle , Protéines foetales/génétique , Fluorouracil/administration et posologie , Régulation de l'expression des gènes tumoraux , Humains , Mâle , Adulte d'âge moyen , Stadification tumorale , Pronostic , Tumeurs du rectum/traitement médicamenteux , Tumeurs du rectum/radiothérapie , Protéines G rho/génétique
19.
J Cell Physiol ; 226(5): 1204-13, 2011 May.
Article de Anglais | MEDLINE | ID: mdl-20945355

RÉSUMÉ

The therapeutic use of ionizing radiation (e.g., X-rays and γ-rays) needs to inflict minimal damage on non-target tissue. Recent studies have shown that substance P (SP) mediates multiple activities in various cell types, including cell proliferation, anti-apoptotic responses, and inflammatory processes. The present study investigated the effects of SP on γ-irradiated bone marrow stem cells (BMSCs). In mouse bone marrow extracts, SP prolonged activation of Erk1/2 and enhanced Bcl-2 expression, but attenuated the activation of apoptotic molecules (e.g., p38 and cleaved caspase-3) and down-regulated Bax. We also observed that SP-decreased apoptotic cell death and stimulated cell proliferation in γ-irradiated mouse bone marrow tissues through TUNEL assay and PCNA analysis. To determine how SP affects bone marrow stem cell populations, mouse bone marrow cells were isolated and colony-forming unit (CFU) of mesenchymal stem cells (MSCs) and hematopoietic stem cells (HSCs) was estimated. SP-pretreated ones showed higher CFUs of MSC and HSC than untreated ones. Furthermore, when SP was pretreated in cultured human MSC, it significantly decreased apoptotic cells at 48 and 72 h after γ-irradiation. Compared with untreated cells, SP-treated human MSCs showed reduced cleavage of apoptotic molecules such as caspase-8, -9, -3, and poly ADP-ribose polymerase (PARP). Thus, our results suggest that SP alleviates γ-radiation-induced damage to mouse BMSCs and human MSCs via regulation of the apoptotic pathway.


Sujet(s)
Cellules de la moelle osseuse/effets des médicaments et des substances chimiques , Cellules de la moelle osseuse/effets des radiations , Cellules souches hématopoïétiques/effets des médicaments et des substances chimiques , Cellules souches hématopoïétiques/effets des radiations , Cellules souches mésenchymateuses/effets des médicaments et des substances chimiques , Cellules souches mésenchymateuses/effets des radiations , Substance P/administration et posologie , Irradiation corporelle totale , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/effets des radiations , Protéines régulatrices de l'apoptose/métabolisme , Cellules de la moelle osseuse/métabolisme , Prolifération cellulaire/effets des médicaments et des substances chimiques , Prolifération cellulaire/effets des radiations , Cellules cultivées , Test clonogénique , Femelle , Cellules souches hématopoïétiques/métabolisme , Humains , Méthode TUNEL , Injections péritoneales , Cellules souches mésenchymateuses/métabolisme , Souris , Souris de lignée C57BL , Antigène nucléaire de prolifération cellulaire/métabolisme , Transduction du signal/effets des médicaments et des substances chimiques , Transduction du signal/effets des radiations , Facteurs temps
20.
Wound Repair Regen ; 17(2): 216-23, 2009.
Article de Anglais | MEDLINE | ID: mdl-19320890

RÉSUMÉ

Radiation therapy causes varying degrees of damage to biological systems. Many groups are investigating the mechanism underlying radiation-induced cellular damage but there are limited therapeutic solutions for affected patients. Recent studies show that substance P (SP) participates in cell proliferation. In the present study, we characterized the mechanism underlying SP-induced cellular signaling in radiation-induced damage of the intestine. Exposure of Caco-2 cells to SP increases cell proliferation and Erk phosphorylation in a time- and dose-dependent manner. The proliferation of cells exposed to gamma-irradiation is also stimulated by exposure to SP, a phenomenon that may result from inhibition of apoptosis because SP activates Akt and inhibits the cleavage of caspase-3. The effect of SP on cell proliferation and protection was confirmed by investigations in mice. Proliferating cell nuclear antigen staining shows that cell proliferation in radiation-damaged mouse intestine increases significantly upon exposure to SP. Furthermore, terminal deoxynucleotidyl transferase-mediated dUTP-fluorescein nick end labeling assay reveals fewer cells stained in SP-treated mice compared with untreated controls. These findings show the potential for SP-induced acceleration of intestinal wound healing and reveal that the mechanism underlying this process involves activation of Erk and Akt and inhibition of caspase-3 cleavage.


Sujet(s)
Cellules Caco-2/effets des radiations , Modèles animaux de maladie humaine , Intestins/traumatismes , Lésions radiques expérimentales , Substance P/physiologie , Cicatrisation de plaie/physiologie , Animaux , Apoptose/physiologie , Technique de Western , Cellules Caco-2/physiologie , Prolifération cellulaire/effets des radiations , Survie cellulaire , Humains , Immunohistochimie , Méthode TUNEL , Mâle , Souris , Souris de lignée C57BL , Agents neuromédiateurs/physiologie , Lésions radiques expérimentales/anatomopathologie , Lésions radiques expérimentales/physiopathologie , Facteurs temps , Irradiation corporelle totale/effets indésirables
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE