Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 131
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Angew Chem Int Ed Engl ; 63(28): e202401333, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38670936

ABSTRACT

The poor electrocatalytic stability and rapid deactivation of metal electrocatalysts are always present in the electrocatalytic conversion of carbon dioxide (CO2) due to the harsh reduction condition. Herein, we demonstrate the controllable dispersion of ultrafine bismuth nanoparticles among the hollow carbon shell (Bi@C-700-4) simply by a thermal-driven diffusion process. The confinement effect of nitrogen-doped carbon matrix is able to low the surface energy of bismuth nanoparticles against the easy aggregation commonly observed for the thermal treatment. On the basis of the synergistic effect and confinement effect between bismuth nanoparticles and carbon matrix, the highly dispersed active sites render the obviously improved electrocatalytic activity and stability for CO2 reduction into formate. The in situ experimental observations on the reduction process and theoretical calculations reveal that the incorporation of bismuth nanoparticles with nitrogen-doped carbon matrix would promote the activation of CO2 and the easy formation of key intermediate (*OCHO), thus leading the enhanced electrocatalytic activity, with a Faradaic Efficiency (FE) of formate about 94.8 % and the long-time stability. Furthermore, the coupling of an anode for 5-hydroxymethylfurfural oxidation reaction (HMFOR) in solar-driven system renders the high 2,5-furandicarboxylic acid (FDCA) yield of 81.2 %, presenting the impressive solar-to-fuel conversion.

2.
Mol Cancer ; 22(1): 199, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38062470

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is one of the most threatening tumors in the world, and chemotherapy remains dominant in the treatment of metastatic CRC (mCRC) patients. The purpose of this study was to develop a biomarker panel to predict the response of the first line chemotherapy in mCRC patients. METHODS: Totally 190 mCRC patients treated with FOLFOX or XEOLX chemotherapy in 3 different institutions were included. We extracted the plasma extracellular vesicle (EV) RNA, performed RNA sequencing, constructed a model and generated a signature through shrinking the number of variables by the random forest algorithm and the least absolute shrinkage and selection operator (LASSO) algorithm in the training cohort (n = 80). We validated it in an internal validation cohort (n = 62) and a prospective external validation cohort (n = 48). RESULTS: We established a signature consisted of 22 EV RNAs which could identify responders, and the area under the receiver operating characteristic curve (AUC) values was 0.986, 0.821, and 0.816 in the training, internal validation, and external validation cohort respectively. The signature could also identify the progression-free survival (PFS) and overall survival (OS). Besides, we constructed a 7-gene signature which could predict tumor response to first-line oxaliplatin-containing chemotherapy and simultaneously resistance to second-line irinotecan-containing chemotherapy. CONCLUSIONS: The study was first to develop a signature of EV-derived RNAs to predict the response of the first line chemotherapy in mCRC with high accuracy using a non-invasive approach, indicating that the signature could help to select the optimal regimen for mCRC patients.


Subject(s)
Cell-Free Nucleic Acids , Colonic Neoplasms , Colorectal Neoplasms , Extracellular Vesicles , Rectal Neoplasms , Humans , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Bevacizumab/therapeutic use , Prospective Studies , Cell-Free Nucleic Acids/genetics , Fluorouracil/therapeutic use , Leucovorin/therapeutic use , RNA , Liquid Biopsy , Extracellular Vesicles/genetics
3.
Int J Cancer ; 152(4): 749-760, 2023 02 15.
Article in English | MEDLINE | ID: mdl-36121651

ABSTRACT

First-line chemotherapy for advanced/metastatic human epidermal growth factor receptor 2 (HER2)-negative gastric/gastroesophageal junction cancer (GC/GEJC) has poor median overall survival (OS; <1 year). We report efficacy and safety results from Chinese patients in the phase III global CheckMate 649 study of nivolumab plus chemotherapy vs chemotherapy for the first-line treatment of GC/GEJC/esophageal adenocarcinoma (EAC). Chinese patients with previously untreated advanced or metastatic GC/GEJC/EAC were randomized to receive nivolumab (360 mg Q3W or 240 mg Q2W) plus chemotherapy (XELOX [capecitabine and oxaliplatin] Q3W or FOLFOX [oxaliplatin, leucovorin and 5-fluorouracil] Q2W), nivolumab plus ipilimumab (not reported) or chemotherapy alone. OS, blinded independent central review-assessed progression-free survival (PFS), objective response rate (ORR), duration of response (DOR) and safety are reported. Of 1581 patients enrolled and randomized, 208 were Chinese. In these patients, nivolumab plus chemotherapy resulted in clinically meaningful improvement in median OS (14.3 vs 10.2 months; HR 0.61 [95% CI: 0.44-0.85]), median PFS (8.3 vs 5.6 months; HR 0.57 [95% CI: 0.40-0.80]), ORR (66% vs 45%) and median DOR (12.2 vs 5.6 months) vs chemotherapy, respectively. The safety profile was acceptable, with no new safety signals observed. Consistent with results from the global primary analysis of CheckMate 649, nivolumab plus chemotherapy demonstrated a clinically meaningful improvement in OS and PFS and higher response rate vs chemotherapy and an acceptable safety profile in Chinese patients. Nivolumab plus chemotherapy represents a new standard first-line treatment for Chinese patients with non-HER2-positive advanced GC/GEJC/EAC.


Subject(s)
Adenocarcinoma , Esophageal Neoplasms , Stomach Neoplasms , Humans , Nivolumab/therapeutic use , Oxaliplatin/therapeutic use , East Asian People , Esophagogastric Junction/pathology , Adenocarcinoma/pathology , Stomach Neoplasms/pathology , Esophageal Neoplasms/pathology , Ipilimumab/therapeutic use , Antineoplastic Combined Chemotherapy Protocols
4.
BMC Med ; 21(1): 173, 2023 05 05.
Article in English | MEDLINE | ID: mdl-37147645

ABSTRACT

BACKGROUND: Apatinib, a highly selective VEGFR2 inhibitor, significantly improved efficacy versus placebo as a third- and later-line treatment for advanced gastric cancer in phase 2 and 3 trials. This prospective, single-arm, multicenter phase IV AHEAD study was conducted to verify the safety and efficacy of apatinib in patients with advanced or metastatic gastric or gastroesophageal adenocarcinoma after at least two lines of systematic therapy in clinical practice settings. METHODS: Patients with advanced gastric cancer who had previously failed at least two lines of chemotherapy received oral apatinib until disease progression, death or unacceptable toxicity. The primary endpoint was safety. The secondary endpoints included objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS) and overall survival (OS). Adverse events were summarized by the incidence rate. Median OS and PFS were estimated using the Kaplan-Meier method. ORR, DCR, OS at 3 and 6 months, and PFS at 3 and 6 months were calculated, and their 95% CIs were estimated according to the Clopper-Pearson method. RESULTS: Between May 2015 and November 2019, a total of 2004 patients were enrolled, and 1999 patients who received at least one dose of apatinib were assessed for safety. In the safety population, 87.9% of patients experienced treatment-related adverse events (TRAEs), with the most common hypertension (45.2%), proteinuria (26.5%), and white blood cell count decreased (25.3%). Additionally, 51% of patients experienced grade ≥ 3 TRAEs. Fatal TRAEs occurred in 57 (2.9%) patients. No new safety concerns were reported. Among the 2004 patients included in the intention-to-treat population, the ORR was 4.4% (95% CI, 3.6-5.4%), and DCR was 35.8% (95% CI, 33.7-38.0%). The median PFS was 2.7 months (95% CI 2.2-2.8), and the median OS was 5.8 months (95% CI 5.4-6.1). CONCLUSIONS: The findings in the AHEAD study confirmed the acceptable and manageable safety profile and clinical benefit of apatinib in patients with advanced gastric cancer as a third- or later-line of treatment. TRIAL REGISTRATION: This study was registered with ClinicalTrials.gov NCT02426034. Registration date was April 24, 2015.


Subject(s)
Adenocarcinoma , Antineoplastic Agents , Stomach Neoplasms , Humans , Antineoplastic Agents/adverse effects , Stomach Neoplasms/drug therapy , Prospective Studies , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Esophagogastric Junction/pathology
5.
Future Oncol ; 17(15): 1923-1931, 2021 May.
Article in English | MEDLINE | ID: mdl-33563040

ABSTRACT

Aim: To assess whether the survival benefit of fruquintinib is quality-adjusted. Materials & methods: Data of 416 metastatic colorectal cancer patients from the Phase III FRESCO trial were used. The Quality-adjusted Time Without Symptoms or Toxicity (Q-TWiST) analysis assessed the quality-adjusted survival benefit of fruquintinib versus placebo, accounting for freedom from symptomatic disease and from severe side effects of treatment. Results: Fruquintinib significantly improved patients' Q-TWiST (difference: 2.23 [1.41, 3.04] months) versus placebo. The Q-TWiST gain was 28.3% in the base case and ranged from 16.7 to 39.9% in the threshold analysis, favoring fruquintinib. The Q-TWiST benefit was observed in fruquintinib-treated patients regardless of prior targeted therapy. Conclusion: Fruquintinib provides a clinically meaningful quality-adjusted survival benefit versus placebo as a third-line treatment for metastatic colorectal cancer patients.


Lay abstract The objective of the study was to assess the benefit of fruqintinib, a chemotherapy drug for patients with metastatic colorectal cancer (mCRC) who do not respond well to previous chemotherapy. The study considered both the time of survival and the quality of life after patients received fruqintinib. In measuring patients' quality of life, the study assessed the time that was free from cancer symptoms and any severe side effects from treatment. The study used data obtained from a Phase III clinical trial, FRESCO, which included 416 mCRC patients receiving fruqintinib or placebo. The results showed that fruqintinib significantly extended patients' symptom-free and side effects-free survival time by approximately 2 months and 5 days. Fruqintinib was 16.7­39.9% more effective than placebo in extending mCRC patients' high-quality life, regardless of prior targeted therapy.


Subject(s)
Benzofurans/administration & dosage , Cancer Survivors/statistics & numerical data , Colorectal Neoplasms/drug therapy , Quality-Adjusted Life Years , Quinazolines/administration & dosage , Aged , Benzofurans/adverse effects , Clinical Trials, Phase III as Topic , Colorectal Neoplasms/mortality , Female , Humans , Male , Middle Aged , Placebos/administration & dosage , Placebos/adverse effects , Quinazolines/adverse effects , Randomized Controlled Trials as Topic , Survival Analysis
6.
Future Oncol ; 17(11): 1339-1350, 2021 Apr.
Article in English | MEDLINE | ID: mdl-33325251

ABSTRACT

Background: FRESCO study demonstrated efficacy and safety of fruquintinib in metastatic colorectal cancer patients. Impact of prior targeted therapy (PTT) on efficacy and safety of fruquintinib was evaluated. Materials & methods: In this subgroup analysis of FRESCO trial, patients were divided into PTT and non-PTT subgroups, and efficacy and safety of fruquintinib were assessed, respectively. Results: In non-PTT subgroup, fruquintinib significantly prolonged overall survival (OS) and progression-free survival (PFS) of patients compared with placebo. In PTT subgroup, the median OS and PFS of patients in fruquintinib arm was significantly higher than those in placebo. Treatment-emergent adverse events (TEAEs) rates were similar in both subgroups. Conclusion: Fruquintinib demonstrated clinically meaningful improvement in OS, PFS, objective response rate, and disease control rate with manageable TEAEs in both subgroups. Clinical trial registration: NCT02314819 (ClinicalTrials.gov).


Lay abstract In this analysis of the FRESCO trial, we evaluated the efficacy and safety of fruquintinib in two different groups of patients (subgroups) with metastatic colorectal cancer - patients who received prior targeted therapy (PTT) and patients who did not (non-PTT). Of the 278 patients treated with fruquintinib, 111 patients received PTT. Patients treated with fruquintinib had longer overall survival and it took longer for their disease to worsen in both PTT and non-PTT subgroups compared with placebo. Patients in both subgroups treated with fruquintinib showed measurable reduction in their tumor size and disease control with similar side effects in patients of both the subgroups. These results suggest that fruquintinib is safe and effective in patients with metastatic colorectal cancer in both subgroups.


Subject(s)
Benzofurans/therapeutic use , Colorectal Neoplasms/drug therapy , ErbB Receptors/antagonists & inhibitors , Quinazolines/therapeutic use , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Aged , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Double-Blind Method , Female , Humans , Male , Middle Aged , Progression-Free Survival , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Survival Rate , Treatment Outcome
7.
Acta Biochim Biophys Sin (Shanghai) ; 53(5): 547-557, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33693450

ABSTRACT

The prognostic significance of c-MET in gastric cancer (GC) remains uncertain. In the present study, we examined the amplification, expression, and the prognostic value of c-MET, human epidermal growth factor receptor 2 (HER2), and programmed cell death 1 ligand 1 (PDL1), together with the correlations among them in a large cohort of Chinese samples. A total of 444 patients were included. The immunohistochemistry (IHC) and the dual-color silver in situ hybridization (SISH) were performed to examine their expression and amplification. Univariate and multivariate analyses were performed by the Cox proportional hazard regression model, and survival curves were estimated by the Kaplan-Meier method. The positivity determined by IHC of c-MET was 24.8%, and the MET amplification rate was 2.3%. The positivity rates of HER2 and PDL1 were 8% and 34.7%, respectively. PDL1 expression had a significantly positive association with c-MET expression. c-MET positivity played a significant prognostic role in disease-free survival (DFS) (P = 0.032). Patients with mesenchymal-epithelial transition (MET) amplification had significantly poorer prognosis on both DFS and overall survival (OS). Subgroup analysis showed that in HER2-negative patients, but not in HER2-positive patients, MET-positive patients had significantly worse DFS (P = 0.000) and OS (P = 0.006). c-MET regulated the expression of PDL1 through an AKT-dependent pathway. c-MET inhibitor enhanced the T-cell killing ability and increased the efficacy of PD1 antibody. c-MET was found to be an independent prognostic factor for DFS of GC patients. A combination of c-MET inhibitors and PD1 antibodies could enhance the killing capacity of T cells, providing a preliminary basis for the clinical research on the same combination in GC treatment.


Subject(s)
B7-H1 Antigen , Gene Amplification , Gene Expression Regulation, Neoplastic , Proto-Oncogene Proteins c-met , Stomach Neoplasms , Up-Regulation , Aged , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/genetics , Disease-Free Survival , Female , Follow-Up Studies , Humans , Male , Middle Aged , Proto-Oncogene Proteins c-met/genetics , Proto-Oncogene Proteins c-met/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/mortality , Survival Rate
8.
J Cell Mol Med ; 24(21): 12550-12559, 2020 11.
Article in English | MEDLINE | ID: mdl-32888389

ABSTRACT

The solute carrier family 52 member 3 (SLC52A3) gene encodes riboflavin transporter protein which is essential to maintain mitochondrial function in cells. In our research, we found that SLC52A3 rs13042395 C > T variation was significantly associated with poor survival in a 926 Chinese gastric cancer (GCa) patients cohort (CC/CT genotype versus TT genotype, HR = 0.57, 95%CI (0.40-0.82), log-rank P = 0.015). The SLC52A3 rs13042395 C > T change led to its increased mRNA expression according to expression quantitative trait loci analysis (P = 0.0029). In vitro, it was revealed that rs13042395 C allele had higher binding affinity to inhibitory transcription factor Meis homeobox 1 (MEIS1) compared with T allele, knock-down of MEIS1 could up-regulate SLC52A3, and overexpression of SLC52A3 contributed to the increased ability of proliferation, colony formation, migration and invasion in GCa cells. Subsequently, the bioinformatics analysis combined with experiments in vitro suggested that Gap junction protein alpha 1 (GJA1) was the downstream effector of SLC52A3, SLC52A3 may promote the GCa cells aggressiveness by down-regulating the GJA1 expression. Overall, SLC52A3 genetic variant rs13042395 C > T change was associated with poorer survival in Chinese GCa patients and increased SLC52A3 expression by interaction with MEIS1. SLC52A3 promoted the GCa cells aggressiveness by down-regulating the GJA1 expression.


Subject(s)
Asian People/genetics , Esophageal Neoplasms/genetics , Genetic Predisposition to Disease , Membrane Transport Proteins/genetics , Polymorphism, Single Nucleotide/genetics , Alleles , Cell Line, Tumor , Connexin 43/genetics , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Myeloid Ecotropic Viral Integration Site 1 Protein/metabolism , Phenotype , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis
9.
Jpn J Clin Oncol ; 50(3): 261-269, 2020 Mar 09.
Article in English | MEDLINE | ID: mdl-31868876

ABSTRACT

PURPOSE: Nomograms are intuitive tools for individualized cancer prognosis. We sought to develop a clinical nomogram for prediction of overall survival and cancer-specific survival for patients with colorectal cancer. METHODS: Patients with colorectal cancer diagnosed between 1988 and 2006 and those who underwent surgery were retrieved from the Surveillance, Epidemiology, and End Results database and randomly divided into the training (n = 119 797) and validation (n = 119 797) cohorts. Log-rank and multivariate Cox regression analyses were used in our analysis. To find out death from other cancer causes and non-cancer causes, a competing-risks model was used, based on which we integrated these significant prognostic factors into nomograms and subjected the nomograms to bootstrap internal validation and to external validation. RESULTS: The 1-, 3-, 5- and 10-year probabilities of overall survival in patients of colorectal cancer after surgery intervention were 83.04, 65.54, 54.79 and 38.62%, respectively. The 1-, 3-, 5- and 10-year cancer-specific survival was 87.36, 73.44, 66.22 and 59.11%, respectively. Nine independent prognostic factors for overall survival and nine independent prognostic factors for cancer specific survival were included to build the nomograms. Internal and external validation CI indexes of overall survival were 0.722 and 0.721, and those of cancer-specific survival were 0.765 and 0.766, which was satisfactory. CONCLUSIONS: Nomograms for prediction of overall survival and cancer-specific survival of patients with colorectal cancer. Performance of the model was excellent. This practical prognostic model may help clinicians in decision-making and design of clinical studies.


Subject(s)
Colorectal Neoplasms/mortality , Models, Statistical , Nomograms , Aged , Colorectal Neoplasms/therapy , Databases, Factual , Female , Humans , Male , Middle Aged , Multivariate Analysis , Prognosis , Reproducibility of Results , Survival Analysis
10.
Acta Biochim Biophys Sin (Shanghai) ; 52(3): 259-267, 2020 Mar 18.
Article in English | MEDLINE | ID: mdl-32147678

ABSTRACT

Tumor metastasis is the main cause of treatment failure and death in patients with late stage of gastric cancer (GC). Studies showed that microRNAs (miRNAs) are important regulators in the process of tumor metastasis. In this study, we used miRNA array analysis to search for metastasis-associated miRNAs in primary and matched metastasis tissues of patients with GC and found that miR-345-5p (miR-345) was significantly higher in primary sites. Decreased expression of miR-345 was observed in GC tissues and cell lines, which was correlated with aggressive stage and grade. Patients with a higher level of miR-345 had a better prognosis. miR-345 could inhibit the migration and spheroid formation abilities in GC cell lines in transwell assay and spheroid formation assay. RNA sequencing and bioinformatics analysis revealed that miR-345 downregulated the epidermal growth factor receptor pathway substrate 8 (EPS8) and its downstream Rac1 signaling. Mechanistically, we confirmed that miR-345 could target EPS8 by directly binding to its 3' untranslated region by luciferase reporter assay. Further rescue assay showed that the ability of miR-345 in inhibiting the migration, stem-like cell phenotype, and epithelial-mesenchymal transition (EMT) in GC was partly dependent on targeting EPS8. In conclusion, miR-345 plays an inhibitory role in GC metastasis through inhibiting cell migration, EMT, and cancer stem cell phenotype via inactivation of Rac1 signaling by targeting EPS8, which provides the potential therapeutic and predictive value of miR-345 in GC.


Subject(s)
Adaptor Proteins, Signal Transducing/metabolism , MicroRNAs/metabolism , Stomach Neoplasms/metabolism , rac1 GTP-Binding Protein/metabolism , 3' Untranslated Regions , Adaptor Proteins, Signal Transducing/genetics , Adult , Aged , Aged, 80 and over , Cell Line, Tumor , Cell Movement/physiology , Down-Regulation , Epithelial-Mesenchymal Transition , Female , Humans , Male , MicroRNAs/genetics , Middle Aged , Neoplasm Metastasis , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Prognosis , RNA Interference , Signal Transduction , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , rac1 GTP-Binding Protein/genetics
11.
Int J Cancer ; 144(2): 251-262, 2019 01 15.
Article in English | MEDLINE | ID: mdl-29978580

ABSTRACT

We previously reported that some single nucleotide polymorphisms (SNPs) of candidate genes involved in the MTOR complex1 (MTORC1) were associated with risk of gastric cancer (GCa). In the present study, we further evaluated associations of eight potentially functional SNPs of MTOR, MLST8 and RPTOR with survival of 1002 GCa patients and also investigated molecular mechanisms underlying such associations. Specifically, we found that the MTOR rs2536 C allele at the microRNA binding site was independently associated with a 26% reduction of death risk (HR = 0.74, 95% CI = 0.57-0.96, p = 0.022). The results remained noteworthy with a prior false positive probability of 0.1. Genotype-phenotype correlation analysis in 144 patients' adjacent normal gastric tissue samples revealed that the MTOR expression levels were lower in rs2536 TC/CC carriers than that in wild-type TT carriers (p = 0.043). Dual luciferase assays revealed that the rs2536 C allele had a higher binding affinity to microRNA-150, leading to a decreased transcriptional activity of MTOR, compared to the rs2536 T allele. Further functional analysis revealed that MTOR knockdown by small interference RNA impaired proliferation, migration, and invasion ability in GCa cell lines. In conclusion, The MTOR rs2536 T > C change may be a biomarker for survival of Chinese GCa patients, likely by modulating microRNA-induced gene expression silencing. Additional studies are needed to validate our findings.


Subject(s)
Stomach Neoplasms/genetics , TOR Serine-Threonine Kinases/genetics , Adult , Aged , Asian People/genetics , Biomarkers, Tumor/genetics , Female , Genetic Association Studies , Genotype , Humans , Male , Middle Aged , Polymorphism, Single Nucleotide , Stomach Neoplasms/mortality
12.
Jpn J Clin Oncol ; 49(5): 438-446, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30924498

ABSTRACT

BACKGROUND: Surgical resection of patients with resectable Stage IV colorectal cancer (CRC) is regarded as first choice if possible. However, its influence on overall survival (OS) has not been thoroughly explored. In this study, we aimed to construct nomograms to help predict 1-, 3- and 5-year OS rate and colorectal cancer-specific survival (CCSS) rate. METHODS: A total of 2996 cases who underwent primary and metastatic resection were selected in the study from surveillance, epidemiology and end results (SEER) database. About 48 Stage IV CRC patients after resection from the Fudan University Shanghai Cancer Center (FUSCC) were assigned as an independent external validation group. Log-rank and multivariate Cox regression analysis were used. The competing-risks model was used to estimate the cumulative incidence of death. Nomograms were built for prediction of OS and CCSS after surgical resection in patients with Stage IV CRC. RESULTS: The 1-, 3- and 5-year probabilities of OS were 76.6%, 41.4% and 23.2%, respectively. The 1-, 3- and 5-year colorectal cumulative incidence of death were 23.0%, 54.9% and 71.3%, respectively. The calibration curves for probability of 1-, 3- and 5-year OS and CCSS showed optimal agreement between nomogram prediction and actual observation, and the Harrell's C-indexes for the nomograms to predict OS and CCSS were 0.662 and 0.650, respectively. For FUSCC validation set, the C-index for this model to predict OS was 0.657. CONCLUSION: Nomograms for prediction of OS and CCSS of patients with Stage IV CRC who underwent primary and metastatic resection were built. Performance of the model was excellent. These nomograms may be helpful for patients and physicians when making a decision.


Subject(s)
Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Nomograms , Adolescent , Adult , Aged , Calibration , Female , Humans , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prognosis , Risk Factors , Young Adult
13.
J Environ Manage ; 251: 109592, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31569022

ABSTRACT

As a way to coordinate the interests of multi-government and solve the problem of transboundary water pollution, watershed ecological compensation system has been promoted in China. It is necessary to understand the influencing factors of watershed ecological compensation from the perspective of how interactions occur between different governments. This paper analyses the interaction among upstream governments, downstream governments and the central government in the Eastern Route of South-to-North Water Transfer Project, using evolutionary game theory. In particular, how ecological benefits are distributed between upstream and downstream governments is analyzed. Simultaneously, numerical simulation is used to analyze the effects of influencing factors on governments' behaviors. The results show that: (1) the initial willingness of governments to corporate affect their final behaviors; (2) upstream and downstream governments cannot spontaneously cooperate to implement watershed ecological compensation system without supervision of the central government; (3) opportunity costs only have a significant impact on upstream governments; (4) punishment on downstream governments can effectively influence the behaviors of governments at all levels; (5) high ecological compensation fee improve downstream governments' willness not to pay; (6) upstream governments get about 78% of ecological benefits due to the implementation of watershed ecological compensation system.


Subject(s)
Ecology , Government , China , Ecosystem , Game Theory
14.
Cancer Sci ; 109(3): 732-740, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29325230

ABSTRACT

Growing evidence suggests that protocadherins (PCDH) play crucial roles in pathogenesis and progression of cancers, including gastric cancer (GC). Protocadherin-8 (PCDH8) was previously reported to be involved in metastasis of GC, but functional studies yielded inconsistent results and the molecular mechanism remained unknown. The present study aimed to explore the clinical relevance, function and molecular mechanism of PCDH8 in GC. Data from the GEPIA and Kaplan-Meier plotter databases showed that high expression of PCDH8 was significantly correlated with poorer prognosis in GC. Ectopic expression of PCDH8 in GC cells promoted invasion and migration in vitro and metastasis in vivo, and knockdown of PCDH8 inhibited invasion and migration in vitro. RNA sequencing followed by gene set enrichment analysis found a remarkable enrichment in the extracellular matrix receptor interaction pathway, with the expression of laminin subunit γ2 (LAMC2) being significantly increased in the PCDH8-overexpressing group. High expression of LAMC2 was significantly correlated to poor prognosis in GC in GEPIA database. Upregulation of LAMC2 following PCDH8 overexpression was further confirmed by immunohistochemistry in liver metastatic lesions of nude mice. To our knowledge, this is the first report of the metastasis-enhancing property and molecular mechanism through upregulation of LAMC2 of PCDH8 in cancer. High expression of PCDH8 could be used as a biomarker for poor prognosis in clinical practice.


Subject(s)
Cadherins/genetics , Cadherins/metabolism , Laminin/genetics , Laminin/metabolism , Stomach Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Movement , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Transplantation , Prognosis , Protocadherins , Stomach Neoplasms/genetics , Survival Analysis , Up-Regulation
15.
Biochem Biophys Res Commun ; 501(4): 1068-1073, 2018 07 02.
Article in English | MEDLINE | ID: mdl-29777710

ABSTRACT

Dysregulation of microRNAs has been demonstrated to be involved in a variety of biological events related to cancer, including proliferation, metastasis, angiogenesis and immune escape. MiR-616-3p is located on the chromosome region 12q13.3, however, its potential role and clinical implications in gastric cancer remain poorly understood. The current study aimed to investigate the potential role of miR-616-3p in gastric cancer. The results showed that miR-616-3p was up-regulated in cancer tissues. Higher expression of miR-616-3p in tumor tissues also predicted poor prognosis. Furthermore, loss- and gain-of-function in vitro revealed that miR-616-3p promoted angiogenesis and EMT in gastric cancer cells. Mechanistically, further analysis demonstrated that the effects of miR-616-3p on metastasis and angiogenesis occurred through the down-regulation of PTEN, a direct target of miR-616-3p. We propose that the restoration of PTEN expression may block miR-616-3p-induced EMT and angiogenesis. Collectively, our findings suggest that the miR-616-3p-PTEN signaling axis might be a potential therapeutic target for gastric cancer.


Subject(s)
Epithelial-Mesenchymal Transition/genetics , MicroRNAs/metabolism , Neovascularization, Pathologic/genetics , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Stomach Neoplasms/blood supply , Stomach Neoplasms/genetics , TOR Serine-Threonine Kinases/metabolism , Base Sequence , Cell Line, Tumor , Cell Movement/genetics , Gene Expression Regulation, Neoplastic , Human Umbilical Vein Endothelial Cells/metabolism , Humans , MicroRNAs/genetics , Neoplasm Invasiveness , Prognosis , Signal Transduction , Stomach Neoplasms/pathology , Up-Regulation/genetics
16.
Acta Biochim Biophys Sin (Shanghai) ; 50(4): 355-361, 2018 Apr 01.
Article in English | MEDLINE | ID: mdl-29534162

ABSTRACT

Mutations in the oncogene BRAF(V600E) are found in ~10% of colorectal cancers (CRCs) and are associated with poor prognosis. However, BRAF(V600E) has a limited response to the small-molecule drug, vemurafenib, a BRAF inhibitor, and BRAF inhibition is thought to cause a feedback activation of EGFR signaling that supports continued proliferation. In this study, we explored the effect of combined use of dabrafenib, a BRAF inhibitor, and cetuximab, an EGFR inhibitor, on BRAF(V600E)-mutant CRC stem cells and its possible mechanisms. Through cell viability analysis, flow cytometry, sphere forming, and western blot analysis, we found that the dabrafenib can synergize with cetuximab to reduce cell viability, induce enhanced apoptotic rates and cell cycle arrest in BRAF(V600E)-mutant HT-29 cells and inhibits stem cell capacities. Further, western blot analysis revealed that PTEN/Src/c-Myc pathway is possibly involved in the synergism between dabrafenib and cetuximab. Overall, our study shows that the combination of dabrafenib and cetuximab results in increased antitumor activity and decreased stem cell capacities in BRAF(V600E)-mutant CRC cells.


Subject(s)
Cetuximab/administration & dosage , Colorectal Neoplasms/genetics , ErbB Receptors/antagonists & inhibitors , Imidazoles/administration & dosage , Neoplastic Stem Cells/drug effects , Oximes/administration & dosage , Proto-Oncogene Proteins B-raf/antagonists & inhibitors , Antineoplastic Agents/administration & dosage , Apoptosis , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Survival , Cetuximab/pharmacology , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Drug Resistance, Neoplasm/genetics , Drug Synergism , ErbB Receptors/metabolism , Flow Cytometry , HT29 Cells , Humans , Imidazoles/pharmacology , Mutation , Neoplasm Transplantation , Neoplastic Stem Cells/cytology , Oximes/pharmacology , Proto-Oncogene Proteins B-raf/metabolism , Proto-Oncogene Proteins c-myc/metabolism , Signal Transduction/drug effects
17.
JAMA ; 319(24): 2486-2496, 2018 06 26.
Article in English | MEDLINE | ID: mdl-29946728

ABSTRACT

Importance: Patients with metastatic colorectal cancer (CRC) have limited effective and tolerable treatment options. Objective: To evaluate the efficacy and safety of oral fruquintinib, a vascular endothelial growth factor receptor (VEGFR) inhibitor, as third-line or later therapy in patients with metastatic CRC. Design, Setting, and Participants: FRESCO (Fruquintinib Efficacy and Safety in 3+ Line Colorectal Cancer Patients) was a randomized, double-blind, placebo-controlled, multicenter (28 hospitals in China), phase 3 clinical trial. From December 2014 to May 2016, screening took place among 519 patients aged 18 to 75 years who had metastatic CRC that progressed after at least 2 lines of chemotherapy but had not received VEGFR inhibitor therapy; 416 met the eligibility criteria and were stratified by prior anti-VEGF therapy and K-ras status. The final date of follow-up was January 17, 2017. Interventions: Patients were randomized in a 2:1 ratio to receive either fruquintinib, 5 mg (n = 278) or placebo (n = 138) orally, once daily for 21 days, followed by 7 days off in 28-day cycles, until disease progression, intolerable toxicity, or study withdrawal. Main Outcomes and Measures: The primary end point was overall survival. Key secondary efficacy endpoints were progression-free survival (time from randomization to disease progression or death), objective response rate (confirmed complete or partial response), and disease control rate (complete or partial response, or stable disease recorded ≥8 weeks postrandomization). Duration of response was also assessed. Safety outcomes included treatment-emergent adverse events. Results: Of the 416 randomized patients (mean age, 54.6 years; 161 [38.7%] women), 404 (97.1%) completed the trial. Median overall survival was significantly prolonged with fruquintinib compared with placebo (9.3 months [95% CI, 8.2-10.5] vs 6.6 months [95% CI, 5.9-8.1]); hazard ratio (HR) for death, 0.65 (95% CI, 0.51-0.83; P < .001). Median progression-free survival was also significantly increased with fruquintinib (3.7 months [95% CI, 3.7-4.6] vs 1.8 months [95% CI, 1.8-1.8] months); HR for progression or death, 0.26 (95% CI, 0.21 to 0.34; P < .001). Grades 3 and 4 treatment-emergent adverse events occurred in 61.2% (170) of patients who received fruquintinib and 19.7% (27) who received placebo. Serious adverse events were reported by 15.5% (43) of patients in the fruquintinib group and 5.8% (8) in the placebo group, with 14.4% (40) of fruquintinib-treated and 5.1% (7) of placebo-treated patients requiring hospitalization. Conclusions and Relevance: Among Chinese patients with metastatic CRC who had tumor progression following at least 2 prior chemotherapy regimens, oral fruquintinib compared with placebo resulted in a statistically significant increase in overall survival. Further research is needed to assess efficacy outside of China. Trial Registration: ClinicalTrials.gov Identifier: NCT02314819.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Benzofurans/administration & dosage , Colorectal Neoplasms/drug therapy , Quinazolines/administration & dosage , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Adult , Aged , Aged, 80 and over , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Benzofurans/adverse effects , China , Colorectal Neoplasms/mortality , Colorectal Neoplasms/pathology , Colorectal Neoplasms/therapy , Combined Modality Therapy , Double-Blind Method , Female , Humans , Intention to Treat Analysis , Male , Middle Aged , Quinazolines/adverse effects , Survival Analysis , Young Adult
18.
Carcinogenesis ; 38(8): 837-846, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28531253

ABSTRACT

Intra-tumor heterogeneity (ITH) is crucial in tumorigenesis and resistance to target therapy. Here, we used mutant-allele tumor heterogeneity (MATH) to measure ITH based on next-generation sequencing data and high MATH was proven as an independent risk prognostic factor in male CRC patients in both a training set of 284 colorectal cancer (CRC) patients with from The Cancer Genome Atlas (TCGA) and a validating set of 187 CRC patients from International Cancer Genome Consortium (ICGC). Further, the genomic pattern according to MATH demonstrated that mutation rates of TP53, IRF5 and KRAS were independently associated with MATH, and the latter two were only significant in male patients. As MATH increased, the fraction of somatic copy number alteration (SCNA) elevated. Moreover, more SCNA events was independently associated with MATH in male than in female. WNT pathway, TGF-ß pathway and DNA repair deficiency was enriched in high MATH group and the latter two showed up only in male patients. In summary, we reveal the gender-related prognostic value of MATH and relevant genomic pattern in CRC. Potential mechanisms are provided and it remains to be proven whether they are drivers of subclone formation and ITH. Taking MATH into consideration in clinical trial might contribute to better therapeutic strategies in CRC with researches added on in the future.


Subject(s)
Carcinogenesis/genetics , Colorectal Neoplasms/genetics , Interferon Regulatory Factors/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Tumor Suppressor Protein p53/genetics , Colorectal Neoplasms/pathology , DNA Copy Number Variations/genetics , Female , Genetic Heterogeneity , Genome, Human , Genomics , Humans , Male , Mutation , Mutation Rate , Prognosis , Sex Characteristics
19.
BMC Cancer ; 17(1): 188, 2017 03 14.
Article in English | MEDLINE | ID: mdl-28288572

ABSTRACT

BACKGROUND: To evaluate the efficacy of cetuximab combined with modified FOLFIRI (mFOLFIRI) as a second-line treatment in metastatic gastric cancer patients and to identify potential biomarkers of clinical outcomes. METHODS: All 61 patients received an initial intravenous (IV) dose of cetuximab (400 mg/m2) and weekly doses (250 mg/m2) thereafter, starting on day 1. On day 2 of each 14-day period, patients received IV irinotecan (180 mg/m2), leucovorin (200 mg/m2), and an IV bolus dose of 5-FU (400 mg/m2) followed by a continuous infusion of 5-FU (2400 mg/m2) for 46 h. The primary endpoint was time-to-progression (TTP). RESULTS: The response rate (RR) was 33.3% among 54 evaluable patients. In the intention-to-treat analysis, median TTP was 4.6 months (95% confidential interval [CI]: 3.6-5.6 months) and median overall survival (OS) was 8.6 months (95% CI: 7.3-9.9 months). In univariate analyses, plasma vascular endothelial growth factor (VEGF) levels were correlated with clinical outcome. In patients with low (≤12.6 pg/ml) and high (>12.6 pg/ml) baseline plasma VEGF levels, RR values were 55.0% and 5.3%, respectively (P = 0.001); median TTP values were 6.9 months and 2.8 months, respectively (P = 0.0005); and median OS values were 12 months and 5 months, respectively (P <0.0001). None of these patients exhibited KRAS, BRAF, or PIK3CA mutations. CONCLUSIONS: Combination therapy comprising cetuximab and mFOLFIRI was well tolerated and active as a second-line treatment for patients with metastatic gastric cancer. Patients with low baseline plasma VEGF levels were associated with better clinical outcomes. TRIAL REGISTRATION: ClinicalTrials.gov. NCT00699881 . Registered 17 June 2008 (retrospectively registered).


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/analysis , Stomach Neoplasms/drug therapy , Vascular Endothelial Growth Factor A/analysis , Adult , Aged , Biomarkers, Tumor/blood , Camptothecin/administration & dosage , Camptothecin/analogs & derivatives , Cetuximab/administration & dosage , Drug Administration Schedule , Female , Fluorouracil/administration & dosage , Humans , Kaplan-Meier Estimate , Leucovorin/administration & dosage , Male , Middle Aged , Nausea/chemically induced , Neoplasm Metastasis , Neutropenia/chemically induced , Stomach Neoplasms/blood , Stomach Neoplasms/pathology , Treatment Outcome , Vascular Endothelial Growth Factor A/blood
20.
Tumour Biol ; 37(2): 1753-62, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26314858

ABSTRACT

This study investigated the associations between genetic polymorphisms of six genes involved in DNA repair, detoxification pathways, and fluoropyrimidine metabolism and clinical outcomes in MGC patients receiving EOF treatment. This retrospective study included 108 Chinese MGC patients receiving EOF as first-line chemotherapy. Nine single nucleotide polymorphisms (SNPs) of six genes (ERCC1 rs2298881, ERCC2 rs13181 and rs1799793, XRCC1 rs25487 and rs25489, GSTP1 rs1695, GSTT1 rs2266637, and MTHFR rs1801133 and rs1801131) were genotyped, and the associations between each SNP and clinical outcome were analyzed. XRCC1 rs25487 A allele was significantly associated with progression disease (PD) to EOF (p = 0.002), and patients with AA genotype had significantly poorer progression-free survival (PFS) (p = 0.001) and overall survival (OS) (p = 0.041) compared with patients with the G allele (GG + GA). ERCC2 rs13181 G allele was significantly associated with PD (p = 0.026), and G carriers (GG + GT) tended to have poorer PFS (p = 0.092) than TT homozygotes. ERCC2 rs1799793 GA genotype was associated with unfavorable PFS (p = 0.034) and a tendency toward poorer OS (p = 0.090) compared with GG homozygotes. Patients were categorized as either good (0 risk factors) or poor risk (≥1 unfavorable SNPs) using a prognostic index based on XRCC1 rs25487 AA, ERCC2 rs13181 (GG + GT), and ERCC2 rs1799793 GA genotypes, with median OS and PFS of 534 days, 281 days (p = 0.009) and 206 days, and 123 days (p < 0.001), respectively. These results suggest that the prognostic index comprising XRCC1 rs25487, ERCC2 rs13181, and rs1799793 polymorphisms may be a useful predictor of clinical outcomes in MGC treated with EOF.


Subject(s)
DNA-Binding Proteins/genetics , Endonucleases/genetics , Glutathione S-Transferase pi/genetics , Glutathione Transferase/genetics , Methylenetetrahydrofolate Reductase (NADPH2)/genetics , Polymorphism, Single Nucleotide/genetics , Stomach Neoplasms/genetics , Xeroderma Pigmentosum Group D Protein/genetics , Alleles , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Asian People/genetics , Disease-Free Survival , Epirubicin/administration & dosage , Female , Fluorouracil/administration & dosage , Genetic Association Studies/methods , Genotype , Humans , Male , Middle Aged , Organoplatinum Compounds/administration & dosage , Oxaliplatin , Prognosis , Retrospective Studies , Stomach Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL