Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
1.
Article in English | MEDLINE | ID: mdl-38305306

ABSTRACT

Digestive system neoplasms are highly heterogeneous and exhibit complex resistance mechanisms that render anti-programmed cell death protein (PD) therapies poorly effective. The tumor microenvironment (TME) plays a pivotal role in tumor development, apart from supplying energy for tumor proliferation and impeding the body's anti-tumor immune response, the TME actively facilitates tumor progression and immune escape via diverse pathways, which include the modulation of heritable gene expression alterations and the intricate interplay with the gut microbiota. In this review, we aim to elucidate the mechanisms underlying drug resistance in digestive tumors, focusing on immune-mediated resistance, microbial crosstalk, metabolism, and epigenetics. We will highlight the unique characteristics of each digestive tumor and emphasize the significance of the tumor immune microenvironment (TIME). Furthermore, we will discuss the current therapeutic strategies that hold promise for combination with cancer immune normalization therapies. This review aims to provide a thorough understanding of the resistance mechanisms in digestive tumors and offer insights into potential therapeutic interventions.

2.
Article in English | MEDLINE | ID: mdl-38275055

ABSTRACT

Tumors of the digestive system are currently one of the leading causes of cancer-related death worldwide. Despite considerable progress in tumor immunotherapy, the prognosis for most patients remains poor. In the tumor microenvironment (TME), tumor cells attain immune escape through immune editing and acquire immune tolerance. The mevalonate pathway and autophagy play important roles in cancer biology, antitumor immunity, and regulation of the TME. In addition, there is metabolic crosstalk between the two pathways. However, their role in promoting immune tolerance in digestive system tumors has not previously been summarized. Therefore, this review focuses on the cancer biology of the mevalonate pathway and autophagy, the regulation of the TME, metabolic crosstalk between the pathways, and the evaluation of their efficacy as targeted inhibitors in clinical tumor immunotherapy.

3.
Cancer Res ; 83(22): 3767-3782, 2023 11 15.
Article in English | MEDLINE | ID: mdl-37646571

ABSTRACT

The chemotherapeutic agent 5-fluorouracil (5-FU) remains the backbone of postoperative adjuvant treatment for gastric cancer. However, fewer than half of patients with gastric cancer benefit from 5-FU-based chemotherapies owing to chemoresistance and limited clinical biomarkers. Here, we identified the SNF2 protein Polo-like kinase 1-interacting checkpoint helicase (PICH) as a predictor of 5-FU chemosensitivity and characterized a transcriptional function of PICH distinct from its role in chromosome separation. PICH formed a transcriptional complex with RNA polymerase II (Pol II) and ATF4 at the CCNA1 promoter in an ATPase-dependent manner. Binding of the PICH complex promoted cyclin A1 transcription and accelerated S-phase progression. Overexpressed PICH impaired 5-FU chemosensitivity in human organoids and patient-derived xenografts. Furthermore, elevated PICH expression was negatively correlated with survival in postoperative patients receiving 5-FU chemotherapy. Together, these findings reveal an ATPase-dependent transcriptional function of PICH that promotes cyclin A1 transcription to drive 5-FU chemoresistance, providing a potential predictive biomarker of 5-FU chemosensitivity for postoperative patients with gastric cancer and prompting further investigation into the transcriptional activity of PICH. SIGNIFICANCE: PICH binds Pol II and ATF4 in an ATPase-dependent manner to form a transcriptional complex that promotes cyclin A1 expression, accelerates S-phase progression, and impairs 5-FU chemosensitivity in gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Cyclin A1 , DNA Helicases/metabolism , Fluorouracil/pharmacology , Adenosine Triphosphatases/therapeutic use , Polo-Like Kinase 1
4.
Gastroenterology ; 164(7): 1232-1247, 2023 06.
Article in English | MEDLINE | ID: mdl-36842710

ABSTRACT

BACKGROUND & AIMS: Although small patient subsets benefit from current targeted strategies or immunotherapy, gemcitabine remains the first-line drug for pancreatic cancer (PC) treatment. However, gemcitabine resistance is widespread and compromises long-term survival. Here, we identified ubiquitin-conjugating enzyme E2T (UBE2T) as a potential therapeutic target to combat gemcitabine resistance in PC. METHODS: Proteomics and metabolomics were combined to examine the effect of UBE2T on pyrimidine metabolism remodeling. Spontaneous PC mice (LSL-KrasG12D/+, LSL-Trp53R172H/+, Pdx1-Cre; KPC) with Ube2t-conditional knockout, organoids, and large-scale clinical samples were used to determine the effect of UBE2T on gemcitabine efficacy. Organoids, patient-derived xenografts (PDX), and KPC mice were used to examine the efficacy of the combination of a UBE2T inhibitor and gemcitabine. RESULTS: Spontaneous PC mice with Ube2t deletion had a marked survival advantage after gemcitabine treatment, and UBE2T levels were positively correlated with gemcitabine resistance in clinical patients. Mechanistically, UBE2T catalyzes ring finger protein 1 (RING1)-mediated ubiquitination of p53 and relieves the transcriptional repression of ribonucleotide reductase subunits M1 and M2, resulting in unrestrained pyrimidine biosynthesis and alleviation of replication stress. Additionally, high-throughput compound library screening using organoids identified pentagalloylglucose (PGG) as a potent UBE2T inhibitor and gemcitabine sensitizer. The combination of gemcitabine and PGG diminished tumor growth in PDX models and prolonged long-term survival in spontaneous PC mice. CONCLUSIONS: Collectively, UBE2T-mediated p53 degradation confers PC gemcitabine resistance by promoting pyrimidine biosynthesis and alleviating replication stress. This study offers an opportunity to improve PC survival by targeting UBE2T and develop a promising gemcitabine sensitizer in clinical translation setting.


Subject(s)
Gemcitabine , Pancreatic Neoplasms , Humans , Mice , Animals , Ubiquitin-Conjugating Enzymes/genetics , Ubiquitin-Conjugating Enzymes/metabolism , Tumor Suppressor Protein p53/genetics , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Disease Models, Animal , Cell Line, Tumor , Pancreatic Neoplasms
5.
Sci Adv ; 8(21): eabn3774, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35613265

ABSTRACT

The diffuse-type gastric cancer (DGC) is a subtype of gastric cancer (GC) associated with low HER2 positivity rate and insensitivity to chemotherapy and immune checkpoint inhibitors. Here, we identify urokinase-type plasminogen activator receptor (uPAR) as a potential therapeutic target for DGC. We have developed a novel anti-uPAR monoclonal antibody, which targets the domains II and III of uPAR and blocks the binding of urokinase-type plasminogen activator to uPAR. We show that the combination of anti-uPAR and anti-Programmed cell death protein 1 (PD-1) remarkably inhibits tumor growth and prolongs survival via multiple mechanisms, using cell line-derived xenograft and patient-derived xenograft mouse models. Furthermore, uPAR chimeric antigen receptor-expressing T cells based on the novel anti-uPAR effectively kill DGC patient-derived organoids and exhibit impressive survival benefit in the established mouse models, especially when combined with PD-1 blockade therapy. Our study provides a new possibility of DGC treatment by targeting uPAR in a unique manner.


Subject(s)
Programmed Cell Death 1 Receptor , Receptors, Urokinase Plasminogen Activator , Stomach Neoplasms , Animals , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/pharmacology , Humans , Mice , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Receptors, Urokinase Plasminogen Activator/antagonists & inhibitors , Receptors, Urokinase Plasminogen Activator/immunology , Signal Transduction , Stomach Neoplasms/drug therapy , Stomach Neoplasms/immunology , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Urokinase-Type Plasminogen Activator/metabolism
6.
BMC Cancer ; 21(1): 1039, 2021 Sep 16.
Article in English | MEDLINE | ID: mdl-34530774

ABSTRACT

BACKGROUND: Pancreatic ductal adenocarcinoma (PDAC) remains a treatment-refractory malignancy with poor prognosis. It is urgent to identify novel and valid biomarkers to predict the progress and prognosis of PDAC. The S100A family have been identified as being involved in cell proliferation, migration and differentiation progression of various cancer types. However, the expression patterns and prognostic values of S100As in PDAC remain to be analyzed. METHODS: We investigated the transcriptional expressions, methylation level and prognostic value of S100As in PDAC patients from the Oncomine, GEPIA2, Linkedomics and cBioPortal databases. Real-time PCR was used to detect the expressions of S100A2/4/6/10/14/16 in four pancreatic cancer cell lines and pancreatic cancer tissues from PDAC patients undergoing surgery. To verify the results further, immunohistochemistry was used to measure the expression of S100A2/4/6/10/14/16 in 43 PDAC patients' tissue samples. The drug relations of S100As were analyzed by using the Drugbank database. RESULTS: The results suggested that, the expression levels of S100A2/4/6/10/14/16 were elevated to PDAC tissues than in normal pancreatic tissues, and the promoter methylation levels of S100A S100A2/4/6/10/14/16 in PDAC (n = 10) were lower compared with normal tissue (n = 184) (P < 0.05). In addition, their expressions were negatively correlated with PDAC patient survival. CONCLUSIONS: Taken together, these results suggest that S100A2/4/6/10/14/16 might be served as prognostic biomarkers for survivals of PDAC patients.


Subject(s)
Adenocarcinoma/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Pancreatic Ductal/metabolism , Pancreatic Neoplasms/metabolism , S100 Proteins/metabolism , Adenocarcinoma/mortality , Annexin A2/metabolism , Calcium-Binding Proteins/metabolism , Carcinoma, Pancreatic Ductal/mortality , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Chemotactic Factors/metabolism , Databases, Genetic , Disease Progression , Humans , Pancreas/metabolism , Pancreatic Neoplasms/mortality , Prognosis , RNA, Messenger/metabolism , S100 Calcium Binding Protein A6/metabolism , S100 Calcium-Binding Protein A4/metabolism , S100 Proteins/genetics , Transcription, Genetic
7.
Nat Commun ; 12(1): 2812, 2021 05 14.
Article in English | MEDLINE | ID: mdl-33990570

ABSTRACT

Trastuzumab is the backbone of HER2-directed gastric cancer therapy, but poor patient response due to insufficient cell sensitivity and drug resistance remains a clinical challenge. Here, we report that HER2 is involved in cell mitotic promotion for tumorigenesis by hyperactivating a crucial HER2-SHCBP1-PLK1 axis that drives trastuzumab sensitivity and is targeted therapeutically. SHCBP1 is an Shc1-binding protein but is detached from scaffold protein Shc1 following HER2 activation. Released SHCBP1 responds to HER2 cascade by translocating into the nucleus following Ser273 phosphorylation, and then contributing to cell mitosis regulation through binding with PLK1 to promote the phosphorylation of the mitotic interactor MISP. Meanwhile, Shc1 is recruited to HER2 for MAPK or PI3K pathways activation. Also, clinical evidence shows that increased SHCBP1 prognosticates a poor response of patients to trastuzumab therapy. Theaflavine-3, 3'-digallate (TFBG) is identified as an inhibitor of the SHCBP1-PLK1 interaction, which is a potential trastuzumab sensitizing agent and, in combination with trastuzumab, is highly efficacious in suppressing HER2-positive gastric cancer growth. These findings suggest an aberrant mitotic HER2-SHCBP1-PLK1 axis underlies trastuzumab sensitivity and offer a new strategy to combat gastric cancer.


Subject(s)
Cell Cycle Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Receptor, ErbB-2/metabolism , Shc Signaling Adaptor Proteins/metabolism , Stomach Neoplasms/drug therapy , Stomach Neoplasms/metabolism , Trastuzumab/pharmacology , Animals , Antineoplastic Agents, Immunological/pharmacology , Biflavonoids/pharmacology , Catechin/analogs & derivatives , Catechin/pharmacology , Cell Cycle Proteins/chemistry , Cell Line, Tumor , Cell Nucleus/metabolism , Drug Resistance, Neoplasm/physiology , Female , Gene Knockdown Techniques , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Male , Mice , Microfilament Proteins/metabolism , Middle Aged , Mitosis/drug effects , Models, Biological , Models, Molecular , Phosphoproteins/metabolism , Prognosis , Protein Interaction Domains and Motifs/drug effects , Protein Serine-Threonine Kinases/chemistry , Proto-Oncogene Proteins/chemistry , Receptor, ErbB-2/antagonists & inhibitors , Shc Signaling Adaptor Proteins/antagonists & inhibitors , Shc Signaling Adaptor Proteins/chemistry , Signal Transduction/drug effects , Stomach Neoplasms/pathology , Xenograft Model Antitumor Assays , Polo-Like Kinase 1
9.
Oncogene ; 40(5): 1027-1042, 2021 02.
Article in English | MEDLINE | ID: mdl-33323973

ABSTRACT

Dysregulation of the Wnt/ß-catenin signaling pathway is critically involved in gastric cancer (GC) progression. However, current Wnt pathway inhibitors being studied in preclinical or clinical settings for other cancers such as colorectal and pancreatic cancers are either too cytotoxic or insufficiently efficacious for GC. Thus, we screened new potent targets from ß-catenin destruction complex associated with GC progression from clinical samples, and found that scaffolding protein RACK1 deficiency plays a significant role in GC progression, but not APC, AXIN, and GSK3ß. Then, we identified its upstream regulator UBE2T which promotes GC progression via hyperactivating the Wnt/ß-catenin signaling pathway through the ubiquitination and degradation of RACK1 at the lysine K172, K225, and K257 residues independent of an E3 ligase. Indeed, UBE2T protein level is negatively associated with prognosis in GC patients, suggesting that UBE2T is a promising target for GC therapy. Furthermore, we identified a novel UBE2T inhibitor, M435-1279, and suggested that M435-1279 acts inhibit the Wnt/ß-catenin signaling pathway hyperactivation through blocking UBE2T-mediated degradation of RACK1, resulting in suppression of GC progression with lower cytotoxicity in the meantime. Overall, we found that increased UBE2T levels promote GC progression via the ubiquitination of RACK1 and identified a novel potent inhibitor providing a balance between growth inhibition and cytotoxicity as well, which offer a new opportunity for the specific GC patients with aberrant Wnt/ß-catenin signaling.


Subject(s)
Neoplasm Proteins/genetics , Receptors for Activated C Kinase/genetics , Stomach Neoplasms/drug therapy , Ubiquitin-Conjugating Enzymes/genetics , beta Catenin/genetics , Animals , Axin Signaling Complex/genetics , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mice , Neoplasm Proteins/antagonists & inhibitors , Receptors for Activated C Kinase/antagonists & inhibitors , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Ubiquitin-Conjugating Enzymes/antagonists & inhibitors , Ubiquitination/drug effects , Wnt Signaling Pathway/genetics , Xenograft Model Antitumor Assays
10.
Int J Oncol ; 56(4): 889-899, 2020 04.
Article in English | MEDLINE | ID: mdl-32319561

ABSTRACT

Gastric cancer (GC) is one of the most frequently diagnosed digestive malignancies and is the third leading cause of cancer­associated death worldwide. Delayed diagnosis and poor prognosis indicate the urgent need for new therapeutic strategies. The success of chimeric antigen receptor (CAR) T­cell therapy for chemotherapy­refractory hematological malignancies has inspired the development of a similar strategy for GC treatment. Although using CAR T­cells against GC is not without difficulty, results from preclinical studies remain encouraging. The current review summarizes relevant preclinical studies and ongoing clinical trials for the use of CAR T­cells for GC treatment and investigates possible toxicities, as well as current clinical experiences and emerging approaches. With a deeper understanding of the tumor microenvironment, novel target epitopes and scientific­technical progress, the potential of CAR T­cell therapy for GC is anticipated in the near future.


Subject(s)
Antigens, Neoplasm/immunology , Drug Evaluation, Preclinical/methods , Immunotherapy, Adoptive/methods , Receptors, Antigen, T-Cell/immunology , Stomach Neoplasms/therapy , T-Lymphocytes/immunology , Tumor Microenvironment/immunology , Clinical Trials as Topic , Humans , Stomach Neoplasms/immunology
11.
BMC Gastroenterol ; 20(1): 100, 2020 Apr 10.
Article in English | MEDLINE | ID: mdl-32276616

ABSTRACT

BACKGROUND: In this study, we aimed to investigate the preoperative serum carcinoembryonic antigen (CEA) in the diagnosis of positive lymph node metastasis (LNM), and to evaluated the relationship between CEA and survival in patients with locally advanced gastric cancer (LAGC). METHODS: The significance of the preoperative serum CEA level for the diagnose of LAGC and prediction of LNM was determined using the receiver operating characteristic (ROC) curve. The areas under the ROC of CEA were compared with those of other tumor markers or imaging examination including CT and MRI. Logistic regression was utilized to identify the risk factors predicting positive LNM. Independent prognosis factors were evaluated using univariate and multivariate COX regression analyses. RESULTS: The ROC curves showed that the AUCs of CEA, CA199, and CA125 for diagnosing LAGC were 0.727, 0.594, and 0.566. When used to predict LNM, the AUC of CEA, CA199 and CA125 were 0.696, 0.531, and 0.588. Logistic regression analysis demonstrated that preoperative serum CEA were significantly associated with positive LNM. On combining imaging examination with CEA, the sensitivity and specificity were 85.3 and 79.4%, respectively, with the AUC equal to 0.853. The combination of CEA and imaging examination preformed the highest levels of AUC and sensitivity for diagnosing LNM, which is significantly higher than using either of them alone. Although patients with abnormal CEA have a poor prognosis, two models of multivariate analysis showed that CEA was not the independent prognosis factor for survival. CONCLUSIONS: CEA can be used to diagnose gastric cancer and determine whether it has LNM. Moreover, combined with CEA could improve the diagnostic sensitivity of imaging examination for lymph node involvement.


Subject(s)
Adenocarcinoma/pathology , Adenocarcinoma/surgery , Carcinoembryonic Antigen/blood , Gastrectomy , Lymphatic Metastasis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Adenocarcinoma/blood , Adenocarcinoma/mortality , Adult , Aged , Area Under Curve , Female , Follow-Up Studies , Humans , Logistic Models , Lymph Node Excision , Male , Middle Aged , Neoplasm Staging , Preoperative Care/methods , Preoperative Period , Prognosis , ROC Curve , Retrospective Studies , Stomach Neoplasms/blood , Stomach Neoplasms/mortality , Survival Analysis
12.
Chin Med J (Engl) ; 133(8): 919-928, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32187050

ABSTRACT

BACKGROUND: Gastric cancer (GC) is one of the most globally prevalent cancers in the world. The pathogenesis of GC has not been fully elucidated, and there still lacks effective targeted therapeutics. The influence of altered kinesin superfamily protein 22 (KIF22) expression in GC progression is still unclearly. The aim of this study was to investigate the KIF22 effects on GC and related mechanisms. METHODS: Gastric carcinoma tissues and matching non-cancerous tissues were collected from patients with GC who have accepted a radical gastrectomy in Lanzhou University Second Hospital from May 2013 to December 2014. The expression of KIF22 was examined in GC of 67 patients and 20 para-carcinoma tissues by immunochemical staining. The relationship between the expression of KIF22 and clinicopathologic characteristics was next investigated in the remaining 52 patients except for 15 patients who did not complete follow-up for 5 years. Cell viability was performed via 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2-H-tetrazolium bromide (MTT) test and colony formation assay in the MGC-803 and BGC-823 GC cells. Cell scratch and trans-well invasion assay was performed to assess migration ability in the MGC-803 and BGC-823 GC cells. Gene set enrichment analysis (GSEA) pathway enrichment analysis was performed to explore the potential functions. Cell cycle was detected by flow cytometry. In addition, the two GC cell lines were used to elucidate the underlying mechanism of KIF22 in GC in vitro via assessing the effects on mitogen-activated protein kinase and extracellular regulated protein kinases (MAPK/ERK) signal transduction pathway-related expressions by Western blotting assays. The differences were compared by t tests, one-way analysis of variance, and Chi-squared tests. RESULTS: The study showed that KIF22 was up-regulated in GC, and KIF22 high expression was significantly related to differentiation degree (χ = 12.842, P = 0.002) and poorly overall survivals. GSEA pathway enrichment analysis showed that KIF22 was correlated with the cell cycle. Silence of KIF22 decreased the ability of the proliferation and migration in gastric cells, induced G1/S phase cell cycle arrest via regulating the MAPK-ERK pathways. CONCLUSIONS: KIF22 protein level was negatively correlated with prognosis. KIF22 knockdown might inhibit proliferation and metastasis of GC cells via the MAPK-ERK signaling pathway.


Subject(s)
Cell Movement/physiology , Cell Proliferation/physiology , DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , Kinesins/metabolism , Stomach Neoplasms/metabolism , Blotting, Western , Cell Cycle/genetics , Cell Cycle/physiology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , DNA-Binding Proteins/genetics , Extracellular Signal-Regulated MAP Kinases/genetics , Female , Flow Cytometry , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/physiology , Humans , Immunohistochemistry , Immunoprecipitation , Kinesins/genetics , MAP Kinase Signaling System/genetics , MAP Kinase Signaling System/physiology , Male , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Stomach Neoplasms/genetics
13.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 45(12): 1490-1498, 2020 Dec 28.
Article in English, Chinese | MEDLINE | ID: mdl-33473008

ABSTRACT

Changes in nuclear morphology are common in malignant tumors, but the underlying molecular mechanisms remain poorly understood. Lamins is involved in supporting nuclear structure, and the expression of Lamins is the molecular basis for nuclear morphological changes during tumor progression. In recent years, the research on the relationship between Lamins and malignant tumors has made great progress. Lamins is of great value in the diagnosis, treatment, and prognosis of various malignant tumors.


Subject(s)
Cell Nucleus , Neoplasms , Humans , Lamins/genetics , Neoplasms/genetics , Prognosis
14.
Int J Surg ; 73: 14-24, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31751791

ABSTRACT

BACKGROUND AND AIM: Superior mesenteric artery (SMA) first approach was a new improvement for pancreaticoduodenectomy (PD), but there is no evidence whether this approach is advantageous to PD. This meta-analysis aimed to determine the effects of the superior mesenteric artery (SMA) first approach on outcomes of pancreaticoduodenectomy (PD). METHODS: Literature searches were conducted on PubMed, The Cochrane Library, EMBASE, Web of Science, Clinical Trials Registry and China Biology Medicine disc. We completed a meta-analysis of the SMA first approach in PD, assessing overall survival, R0 resection, blood loss, postoperative complications, operation time and postoperative stay. The odds ratios and weighted mean differences with 95% confidence intervals (CIs) were pooled. RESULTS: Eighteen studies comprising 1483 participants were included. Patients who received SMA-PD had significantly lower overall complication rate (OR 0.62, 95% CI 0.47 to 0.81, P = 0.001) and less blood loss (WMD -264.84, 95% CI -336.1 to -193.58, P < 0.001). The obviously increased R0 resection rate (OR 2.92, 95% CI 1.72 to 4.96, P < 0.001) and 3-year OS (OR 2.15, 95% CI 1.34 to 3.43, P = 0.001) were found in the SMA-PD group. CONCLUSION: The SMA-PD group had better clinical outcomes, particularly in long-term survival of pancreatic cancer patients; furthermore, the patients acquired superior clinical efficacy via the posterior approach in SMA-PD.


Subject(s)
Mesenteric Artery, Superior/surgery , Pancreatic Neoplasms/surgery , Pancreaticoduodenectomy/methods , Adult , Aged , Aged, 80 and over , Female , Humans , Male , Middle Aged , Pancreaticoduodenectomy/adverse effects , Postoperative Complications/etiology , Treatment Outcome
15.
Fertil Steril ; 112(1): 89-97.e1, 2019 07.
Article in English | MEDLINE | ID: mdl-31277770

ABSTRACT

OBJECTIVE: To evaluate whether intrauterine injection of hCG before embryo transfer can improve IVF-ET outcomes. DESIGN: Meta-analysis. SETTING: Not applicable. PATIENT(S): Infertile women who underwent IVF-ET and received an intrauterine injection of hCG before ET. INTERVENTION(S): Infertile women treated with or without intrauterine hCG injection before ET. MAIN OUTCOME MEASURE(S): The primary outcomes were live birth rate (LBR), ongoing pregnancy rate (OPR), and clinical pregnancy rate (CPR), and the secondary outcomes were implantation rate (IR) and miscarriage rate (MR). Odds ratios with 95% confidence intervals (CIs) and successful ET rates were pooled to determine the effects of hCG on IVF-ET outcomes. RESULT(S): Fifteen randomized controlled trials (RCTs) with a total of 2,763 participants were included. Infertile women in the experimental group (treated with intrauterine hCG injection before ET) exhibited significantly higher LBR (44.89% vs. 29.76%), OPR (48.09% vs. 33.42%), CPR (47.80% vs. 32.78%), and IR (31.64% vs. 22.52%) than those in the control group (intrauterine injection of placebo or no injection). Furthermore, MR was significantly lower (12.45% vs. 18.56%) in the experimental group than in the control group. CONCLUSION(S): The findings of this meta-analysis indicate that intrauterine injection of hCG can improve LBR, OPR, CPR, and IR after IVF-ET cycles. In addition, different timing and dosages of hCG administration may exert different effects on IVT-ET outcomes. Notably, infertile women treated with 500 IU hCG within 15 minutes before ET can achieve optimal IVF-ET outcomes.


Subject(s)
Chorionic Gonadotropin/administration & dosage , Embryo Transfer , Fertility Agents, Female/administration & dosage , Fertilization in Vitro , Infertility, Female/therapy , Chorionic Gonadotropin/adverse effects , Embryo Implantation/drug effects , Embryo Transfer/adverse effects , Female , Fertility/drug effects , Fertility Agents, Female/adverse effects , Fertilization in Vitro/adverse effects , Humans , Infertility, Female/diagnosis , Infertility, Female/physiopathology , Injections , Live Birth , Pregnancy , Pregnancy Complications/etiology , Pregnancy Rate , Randomized Controlled Trials as Topic , Risk Factors , Treatment Outcome
16.
Curr Cancer Drug Targets ; 19(11): 854-862, 2019.
Article in English | MEDLINE | ID: mdl-31250756

ABSTRACT

Src homolog and collagen homolog (Shc) proteins have been identified as adapter proteins associated with cell surface receptors and have been shown to play important roles in signaling and disease. Shcbp1 acts as a Shc SH2-domain binding protein 1 and is involved in the regulation of signaling pathways, such as FGF, NF-κB, MAPK/ERK, PI3K/AKT, TGF-ß1/Smad and ß -catenin signaling. Shcbp1 participates in T cell development, the regulation of downstream signal transduction pathways, and cytokinesis during mitosis and meiosis. In addition, Shcbp1 has been demonstrated to correlate with Burkitt-like lymphoma, breast cancer, lung cancer, gliomas, synovial sarcoma, human hepatocellular carcinoma and other diseases. Shcbp1 may play an important role in tumorigenesis and progression. Accordingly, recent studies are reviewed herein to discuss and interpret the role of Shcbp1 in normal cell proliferation and differentiation, tumorigenesis and progression, as well as its interactions with proteins.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/pathology , Shc Signaling Adaptor Proteins/metabolism , Signal Transduction , Animals , Cell Cycle , Cell Proliferation , Disease Progression , Humans , Mitosis , Neoplasms/genetics , Neoplasms/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...