Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 70
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Lancet ; 402(10408): 1133-1146, 2023 09 30.
Article in English | MEDLINE | ID: mdl-37499670

ABSTRACT

BACKGROUND: Immunotherapy with immune checkpoint inhibitors combined with an anti-angiogenic tyrosine-kinase inhibitor (TKI) has been shown to improve overall survival versus anti-angiogenic therapy alone in advanced solid tumours, but not in hepatocellular carcinoma. Therefore, a clinical study was conducted to compare the efficacy and safety of the anti-PD-1 antibody camrelizumab plus the VEGFR2-targeted TKI rivoceranib (also known as apatinib) versus sorafenib as first-line treatment for unresectable hepatocellular carcinoma. METHODS: This randomised, open-label, international phase 3 trial (CARES-310) was done at 95 study sites across 13 countries and regions worldwide. Patients with unresectable or metastatic hepatocellular carcinoma who had not previously received any systemic treatment were randomly assigned (1:1) to receive either camrelizumab 200 mg intravenously every 2 weeks plus rivoceranib 250 mg orally once daily or sorafenib 400 mg orally twice daily. Randomisation was done via a centralised interactive response system. The primary endpoints were progression-free survival, as assessed by the blinded independent review committee per Response Evaluation Criteria in Solid Tumours version 1.1, and overall survival in the intention-to-treat population. Safety was assessed in all patients who received at least one dose of the study drugs. We report the findings from the prespecified primary analysis for progression-free survival and interim analysis for overall survival. This study is registered with ClinicalTrials.gov (NCT03764293). FINDINGS: Between June 28, 2019, and March 24, 2021, 543 patients were randomly assigned to the camrelizumab-rivoceranib (n=272) or sorafenib (n=271) group. At the primary analysis for progression-free survival (May 10, 2021), median follow-up was 7·8 months (IQR 4·1-10·6). Median progression-free survival was significantly improved with camrelizumab-rivoceranib versus sorafenib (5·6 months [95% CI 5·5-6·3] vs 3·7 months [2·8-3·7]; hazard ratio [HR] 0·52 [95% CI 0·41-0·65]; one-sided p<0·0001). At the interim analysis for overall survival (Feb 8, 2022), median follow-up was 14·5 months (IQR 9·1-18·7). Median overall survival was significantly extended with camrelizumab-rivoceranib versus sorafenib (22·1 months [95% CI 19·1-27·2] vs 15·2 months [13·0-18·5]; HR 0·62 [95% CI 0·49-0·80]; one-sided p<0·0001). The most common grade 3 or 4 treatment-related adverse events were hypertension (102 [38%] of 272 patients in the camrelizumab-rivoceranib group vs 40 [15%] of 269 patients in the sorafenib group), palmar-plantar erythrodysaesthesia syndrome (33 [12%] vs 41 [15%]), increased aspartate aminotransferase (45 [17%] vs 14 [5%]), and increased alanine aminotransferase (35 [13%] vs eight [3%]). Treatment-related serious adverse events were reported in 66 (24%) patients in the camrelizumab-rivoceranib group and 16 (6%) in the sorafenib group. Treatment-related death occurred in two patients: one patient in the camrelizumab-rivoceranib group (ie, multiple organ dysfunction syndrome) and one patient in the sorafenib group (ie, respiratory failure and circulatory collapse). INTERPRETATION: Camrelizumab plus rivoceranib showed a statistically significant and clinically meaningful benefit in progression-free survival and overall survival compared with sorafenib for patients with unresectable hepatocellular carcinoma, presenting as a new and effective first-line treatment option for this population. FUNDING: Jiangsu Hengrui Pharmaceuticals and Elevar Therapeutics.


Subject(s)
Carcinoma, Hepatocellular , Liver Neoplasms , Humans , Carcinoma, Hepatocellular/drug therapy , Sorafenib/therapeutic use , Liver Neoplasms/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use
2.
Cancer Immunol Immunother ; 73(7): 124, 2024 May 10.
Article in English | MEDLINE | ID: mdl-38727837

ABSTRACT

BACKGROUND: The combination of immune checkpoint inhibitors and antiangiogenic agents has been effective in treating multiple cancers. This was further explored in an open-label, multicenter phase 2 basket study (NCT04346381), which evaluated the antitumor activity and safety of camrelizumab (an anti-PD-1 antibody) plus famitinib (a receptor tyrosine kinase inhibitor) in patients with advanced solid tumors. We herein report the findings from the cohort of advanced NSCLC patients who progressed after treatment with platinum-doublet chemotherapy and immunotherapy. METHODS: Eligible patients were enrolled and treated with camrelizumab (200 mg once every 3 weeks via intravenous infusion) and oral famitinib (20 mg once daily). The primary endpoint was the objective response rate (ORR). Secondary endpoints included the disease control rate (DCR), duration of response (DoR), progression-free survival (PFS), overall survival (OS), and safety. RESULTS: Forty patients were enrolled in this cohort, with a median follow-up duration of 11.5 months. Three patients (7.5%) achieved a partial response, and 29 patients (72.5%) achieved stable disease. The ORR and DCR with this combination regimen were 7.5% (95% CI, 1.6-20.4) and 80.0% (95% CI, 64.4-90.9), respectively. The median DoR was 12.1 months (95% CI, 10.3-not reached). The median PFS was 5.4 months (95% CI, 4.1-7.5), and the median OS was 12.1 months (95% CI, 9.1-16.7). The estimated 12-month OS rate was 51.5% (95% CI, 34.9-65.9). The most frequent grade 3 or higher treatment-related adverse events occurring in more than 5% of patients included hypertension (27.5%), palmar-plantar erythrodysesthesia syndrome (10%), decreased neutrophil count (10%), and proteinuria (7.5%). CONCLUSION: Camrelizumab plus famitinib demonstrated favorable benefits in PFS and OS, along with manageable safety profiles, in patients with advanced NSCLC who progressed after platinum-doublet chemotherapy and immunotherapy. This finding warrants further exploration.


Subject(s)
Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Male , Female , Middle Aged , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Aged , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Antibodies, Monoclonal, Humanized/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Adult , Sulfonamides/therapeutic use , Sulfonamides/administration & dosage , Immunotherapy/methods , Indoles , Pyrroles
3.
BMC Bioinformatics ; 23(1): 390, 2022 Sep 27.
Article in English | MEDLINE | ID: mdl-36167487

ABSTRACT

BACKGROUND: Collagen VI family (COL6A) is a major member of extracellular matrix protein. There is accumulating evidence that COL6A is involved in tumorigenesis and tumor progression. In this study, we performed a systematic analysis of COL6A in pan-cancer based on their molecular features and clinical significance. METHODS: Based on updated public databases, we integrated several bioinformatics analysis methods to investigate the expression levels of COL6A as well as the relationship between their expression and patient survival, immune subtypes, tumor microenvironment, stemness scores, drug sensitivity, and DNA methylation. RESULTS: The expression levels of COL6A members varied in different cancers, suggesting their expression was cancer-dependent. Among COL6A members, COL6A1/2/3 were predicted poor prognosis in specific cancers. Furthermore, COL6A1/2/3 expression levels revealed a clear correlation with immune subtypes, and COL6A1/2/3 were associated with tumor purity, that is, gene expression levels were generally higher in tumors with higher stromal scores and immune scores. COL6A1/2/3 had a significantly negative correlation with RNA stemness scores, and meanwhile they were also related to DNA stemness scores in different degrees. In addition, the expression of COL6A1/2/3 was significantly related to drug sensitivity of cancer cells. Finally, our study revealed that COL6A1/2/3 expression was mainly negatively correlated with gene methylation, and the methylation levels showed remarkable differences in various cancers. CONCLUSIONS: These findings highlight both the similarities and differences in the molecular characteristics of COL6A members in pan-cancer, and provide comprehensive insights for further investigation into the mechanism of COL6A.


Subject(s)
Neoplasms , Tumor Microenvironment , Collagen Type VI/genetics , Collagen Type VI/metabolism , DNA Methylation , Humans , Neoplasms/drug therapy , Neoplasms/genetics , RNA/metabolism
4.
Carcinogenesis ; 43(12): 1162-1175, 2022 12 31.
Article in English | MEDLINE | ID: mdl-36194598

ABSTRACT

The Notch1 (Notch1 receptor) and yes-associated protein 1 (YAP1) signaling can regulate breast cancer metastasis. This study aimed at investigating whether and how these two signal pathways crosstalk to promote breast cancer lung metastasis. Here, we show that YAP1 expression was positively correlated with Notch1 in breast cancer according to bioinformatics and experimental validation. Mechanistically, YAP1 with TEA domain transcription factors (TEADs) enhanced Jagged1(JAG1)-Notch1 signaling. Meanwhile, Notch1 promoted YAP1 stability in breast cancer cells by inhibiting the ß-TrCP-mediated degradation, thereby, forming a YAP1- JAG1/Notch1 positive feedback loop in breast cancer. Furthermore, YAP1 enhanced the mammosphere formation and stemness of MDA-MB-231 cells by attenuating the inhibition of the BMP4-SMAD1/5 signaling. In vivo, the YAP1- JAG1/Notch1 positive feedback loop promoted the lung colonization of MDA-MB-231 cells. Our data for the first time indicate that the YAP1-Notch1 positive feedback loop promotes lung metastasis of breast cancer by modulating self-renewal and inhibiting the BMP4-SMAD1/5 signaling.


Subject(s)
Breast Neoplasms , Lung Neoplasms , Humans , Female , Receptor, Notch1/genetics , Receptor, Notch1/metabolism , Breast Neoplasms/pathology , YAP-Signaling Proteins , Feedback , Bone Morphogenetic Protein 4/metabolism , Signal Transduction , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Lung Neoplasms/genetics , Family , Cell Line, Tumor
5.
Lancet Oncol ; 22(7): 977-990, 2021 07.
Article in English | MEDLINE | ID: mdl-34143971

ABSTRACT

BACKGROUND: China has a high burden of hepatocellular carcinoma, and hepatitis B virus (HBV) infection is the main causative factor. Patients with hepatocellular carcinoma have a poor prognosis and a substantial unmet clinical need. The phase 2-3 ORIENT-32 study aimed to assess sintilimab (a PD-1 inhibitor) plus IBI305, a bevacizumab biosimilar, versus sorafenib as a first-line treatment for unresectable HBV-associated hepatocellular carcinoma. METHODS: This randomised, open-label, phase 2-3 study was done at 50 clinical sites in China. Patients aged 18 years or older with histologically or cytologically diagnosed or clinically confirmed unresectable or metastatic hepatocellular carcinoma, no previous systemic treatment, and a baseline Eastern Cooperative Oncology Group (ECOG) performance status of 0 or 1 were eligible for inclusion. In the phase 2 part of the study, patients received intravenous sintilimab (200 mg every 3 weeks) plus intravenous IBI305 (15 mg/kg every 3 weeks). In the phase 3 part, patients were randomly assigned (2:1) to receive either sintilimab plus IBI305 (sintilimab-bevacizumab biosimilar group) or sorafenib (400 mg orally twice daily; sorafenib group), until disease progression or unacceptable toxicity. Randomisation was done using permuted block randomisation, with a block size of six, via an interactive web response system, and stratified by macrovascular invasion or extrahepatic metastasis, baseline α-fetoprotein, and ECOG performance status. The primary endpoint of the phase 2 part of the study was safety, assessed in all patients who received at least one dose of study drug. The co-primary endpoints of the phase 3 part of the study were overall survival and independent radiological review committee (IRRC)-assessed progression-free survival according to Response Evaluation Criteria in Solid Tumors (RECIST) version 1.1 in the intention-to-treat population. The study is registered with ClinicalTrials.gov, NCT03794440. The study is closed to new participants and follow-up is ongoing for long-term outcomes. FINDINGS: Between Feb 11, 2019 and Jan 15, 2020, we enrolled 595 patients: 24 were enrolled directly into the phase 2 safety run-in and 571 were randomly assigned to sintilimab-bevacizumab biosimilar (n=380) or sorafenib (n=191). In the phase 2 part of the trial, 24 patients received at least one dose of the study drug, with an objective response rate of 25·0% (95% CI 9·8-46·7). Based on the preliminary safety and activity data of the phase 2 part, in which grade 3 or worse treatment-related adverse events occurred in seven (29%) of 24 patients, the randomised phase 3 part was started. At data cutoff (Aug 15, 2020), the median follow-up was 10·0 months (IQR 8·5-11·7) in the sintilimab-bevacizumab biosimilar group and 10·0 months (8·4-11·7) in the sorafenib group. Patients in the sintilimab-bevacizumab biosimilar group had a significantly longer IRRC-assessed median progression-free survival (4·6 months [95% CI 4·1-5·7]) than did patients in the sorafenib group (2·8 months [2·7-3·2]; stratified hazard ratio [HR] 0·56, 95% CI 0·46-0·70; p<0·0001). In the first interim analysis of overall survival, sintilimab-bevacizumab biosimilar showed a significantly longer overall survival than did sorafenib (median not reached [95% CI not reached-not reached] vs 10·4 months [8·5-not reached]; HR 0·57, 95% CI 0·43-0·75; p<0·0001). The most common grade 3-4 treatment-emergent adverse events were hypertension (55 [14%] of 380 patients in the sintilimab-bevacizumab biosimilar group vs 11 [6%] of 185 patients in the sorafenib group) and palmar-plantar erythrodysaesthesia syndrome (none vs 22 [12%]). 123 (32%) patients in the sintilimab-bevacizumab biosimilar group and 36 (19%) patients in the sorafenib group had serious adverse events. Treatment-related adverse events that led to death occurred in six (2%) patients in the sintilimab-bevacizumab biosimilar group (one patient with abnormal liver function, one patient with both hepatic failure and gastrointestinal haemorrhage, one patient with interstitial lung disease, one patient with both hepatic faliure and hyperkalemia, one patient with upper gastrointestinal haemorrhage, and one patient with intestinal volvulus) and two (1%) patients in the sorafenib group (one patient with gastrointestinal haemorrhage and one patient with death of unknown cause). INTERPRETATION: Sintilimab plus IBI305 showed a significant overall survival and progression-free survival benefit versus sorafenib in the first-line setting for Chinese patients with unresectable, HBV-associated hepatocellular carcinoma, with an acceptable safety profile. This combination regimen could provide a novel treatment option for such patients. FUNDING: Innovent Biologics. TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Biosimilar Pharmaceuticals/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Liver Neoplasms/drug therapy , Sorafenib/therapeutic use , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Bevacizumab/adverse effects , Biosimilar Pharmaceuticals/adverse effects , Carcinoma, Hepatocellular/mortality , Carcinoma, Hepatocellular/pathology , Carcinoma, Hepatocellular/virology , China , Disease Progression , Female , Hepatitis B/virology , Humans , Liver Neoplasms/mortality , Liver Neoplasms/pathology , Liver Neoplasms/virology , Male , Middle Aged , Progression-Free Survival , Sorafenib/adverse effects , Time Factors , Young Adult
6.
Int J Cancer ; 149(11): 1944-1954, 2021 12 01.
Article in English | MEDLINE | ID: mdl-34309846

ABSTRACT

Biliary tract cancer (BTC) is a highly malignant tumor with limited treatment options and poor prognosis. Our study aimed to evaluate camrelizumab plus oxaliplatin-based chemotherapy as first-line therapy for advanced BTC. In this multicenter, open-label, phase 2 trial conducted in China (ClinicalTrials.gov, NCT03092895), untreated patients with advanced BTC were given camrelizumab (3 mg/kg iv drip injection, every 2 weeks) plus typical FOLFOX4 (Cam-FOLFOX4 group; infusional 5-fluorouracil, leucovorin and oxaliplatin) or GEMOX (Cam-GEMOX group; infusional gemcitabine and oxaliplatin). The primary endpoint was objective response rate (ORR). Ninety-two patients were enrolled: 29 received Cam-FOLFOX4 and 63 received Cam-GEMOX. The confirmed ORR and disease control rate were 16.3% (95% confidence interval [CI] = 9.4-25.5) and 75.0% (95% CI = 64.9-83.4), respectively. Median duration of response was 8.7 months (95% CI = 5.1-not reached). Median progression-free survival and overall survival were 5.3 months (95% CI = 3.7-5.7) and 12.4 months (95% CI = 8.9-16.1), respectively. Grade ≥3 treatment-related adverse events (TRAEs) occurred in 82.8% of patients receiving Cam-FOLFOX4 and in 68.3% receiving Cam-GEMOX, with no unexpected effects observed. Six (6.5%) patients discontinued treatment due to TRAE. Camrelizumab plus FOLFOX4 or GEMOX as first-line treatment was effective and tolerable for Chinese patients with advanced BTC, warranting phase 3 trials.


Subject(s)
Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biliary Tract Neoplasms/drug therapy , Oxaliplatin/therapeutic use , Adult , Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Biliary Tract Neoplasms/pathology , China , Female , Humans , Male , Middle Aged , Oxaliplatin/adverse effects , Progression-Free Survival , Safety , Treatment Outcome
7.
Exp Cell Res ; 392(2): 112034, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32339606

ABSTRACT

Programmed death-ligand 1 (PD-L1) expression on the surface of tumour cells can cause tumour immune evasion. Benefits of combining anti-PD-L1 therapy with nab-paclitaxel in patients with advanced triple-negative breast cancer (TNBC) have been reported. However, some patients cannot tolerate the immune-related adverse effects (irAEs) caused by antibody-based immunotherapy. BRD4 is a member of the bromodomain and extra-terminal domain (BET) family. BRD4 inhibition has shown antitumour effects in many tumours, but its role in TNBC has not been definitively concluded. In particular, the immune regulation of BRD4 in TNBC has been rarely studied. In this study, we used JQ1, a BET inhibitor, and small interfering RNAs (siRNAs) targeting BRD4 to explore the influence of BRD4 on PD-L1 expression in TNBC. The results indicated that BRD4 inhibition suppressed PD-L1 expression and the PD-L1 upregulation induced by interferon-γ (IFN-γ). In the in vivo experiments, we found that JQ1 not only reduced the PD-L1 expression level but also changed the proportions of T lymphocyte subsets in the spleens of tumour-bearing mice, which helped to relieve immunosuppression. Briefly, our study reveals that BRD4 regulates PD-L1 expression and may provide a potential method for blocking the programmed death 1 (PD-1)/PD-L1 immune checkpoint in TNBC.


Subject(s)
Azepines/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Cell Cycle Proteins/antagonists & inhibitors , Gene Expression Regulation, Neoplastic/drug effects , Transcription Factors/antagonists & inhibitors , Triazoles/pharmacology , Triple Negative Breast Neoplasms/pathology , Animals , Apoptosis , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Biomarkers, Tumor/genetics , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Proliferation , Female , Humans , Interferon-gamma/metabolism , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Transcription Factors/genetics , Transcription Factors/metabolism , Triple Negative Breast Neoplasms/genetics , Triple Negative Breast Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
8.
Pharmacology ; 106(9-10): 509-519, 2021.
Article in English | MEDLINE | ID: mdl-34412054

ABSTRACT

INTRODUCTION: Intrahepatic cholangiocarcinoma (ICC), which is difficult to diagnose and is usually fatal due to its late clinical presentation and a lack of effective treatment, has risen over the past decades but without much improvement in prognosis. OBJECTIVE: The study aimed to investigate the role of apatinib that targets vascular endothelial growth factor receptor-2 (VEGFR2) in ICC. METHODS: MTT assays, cell scratch assays, and tube formation assays were used to assess the effect of apatinib on human ICC cell line (HuCCT-1) and RBE cells proliferation, migration, and angiogenic capacity, respectively. Expression of vascular endothelial growth factor (VEGF), VEGFR2, signal transducer and activator of transcription factor 3 (STAT3), pSTAT3, and hypoxia inducible factor 1 subunit alpha (HIF-1α) pathway proteins was assessed using Western blotting and mRNA expression analysis in HuCCT-1 was performed using RT-qPCR assays. The pcDNA 3.1(-)-VEGFR2 and pcDNA 3.1(-)-HIF-1α were transfected into HuCCT-1 and RBE cells using Lipofectamine 2,000 to obtain overexpressed HuCCT-1 and RBE cells. RESULTS: We found that apatinib-inhibited proliferation, migration, and angiogenesis of HuCCT-1 and RBE cells in vitro in a dose-dependent manner. We also proved that apatinib effectively inhibits angiogenesis in tumor cells by blocking the expression of VEGF and VEGFR2 in these cells. In addition, we demonstrated that apatinib regulates the expression of STAT3 phosphorylation by inhibiting VEGFR2. Finally, we showed that apatinib regulates ICC angiogenesis and HIF-1α/VEGF expression via STAT3. CONCLUSIONS: Based on the above findings, we conclude that apatinib inhibits HuCCT-1 and RBE cell proliferation, migration, and tumor angiogenesis by inhibiting the VEGFR2/STAT3/HIF-1α axis signaling pathway. Apatinib can be a promising drug for ICC-targeted molecular therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Bile Duct Neoplasms/pathology , Cholangiocarcinoma/pathology , Neovascularization, Pathologic/pathology , Pyridines/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Hypoxia-Inducible Factor 1/drug effects , Signal Transduction/drug effects , Transcription Factor 3/drug effects , Vascular Endothelial Growth Factor Receptor-2/drug effects
9.
Entropy (Basel) ; 23(9)2021 Aug 31.
Article in English | MEDLINE | ID: mdl-34573757

ABSTRACT

The aim of multi-agent reinforcement learning systems is to provide interacting agents with the ability to collaboratively learn and adapt to the behavior of other agents. Typically, an agent receives its private observations providing a partial view of the true state of the environment. However, in realistic settings, the harsh environment might cause one or more agents to show arbitrarily faulty or malicious behavior, which may suffice to allow the current coordination mechanisms fail. In this paper, we study a practical scenario of multi-agent reinforcement learning systems considering the security issues in the presence of agents with arbitrarily faulty or malicious behavior. The previous state-of-the-art work that coped with extremely noisy environments was designed on the basis that the noise intensity in the environment was known in advance. However, when the noise intensity changes, the existing method has to adjust the configuration of the model to learn in new environments, which limits the practical applications. To overcome these difficulties, we present an Attention-based Fault-Tolerant (FT-Attn) model, which can select not only correct, but also relevant information for each agent at every time step in noisy environments. The multihead attention mechanism enables the agents to learn effective communication policies through experience concurrent with the action policies. Empirical results showed that FT-Attn beats previous state-of-the-art methods in some extremely noisy environments in both cooperative and competitive scenarios, much closer to the upper-bound performance. Furthermore, FT-Attn maintains a more general fault tolerance ability and does not rely on the prior knowledge about the noise intensity of the environment.

10.
J Cell Biochem ; 120(8): 13985-13993, 2019 08.
Article in English | MEDLINE | ID: mdl-30957285

ABSTRACT

Matrix attachment regions (MARs) can enhance transgene expression levels and maintain stability. However, the consensus sequence from MARs and its functional analysis remains to be examined. Here, we assessed a possible consensus sequence from MARs and assessed its activity in stably transfected Chinese hamster ovary (CHO) cells. First, we analyzed the effects of 10 MARs on transfected CHO cells and then analyzed the consensus motifs from these MARs using a bioinformatics method. The consensus sequence was synthesized and cloned upstream or downstream of the eukaryotic vector. The constructs were transfected into CHO cells and the expression levels and stability of enhanced green fluorescent protein were detected by flow cytometry. The results indicated that eight of the ten MARs increased transgene expression in transfected CHO cells. Three consensus motifs were found after bioinformatics analyses. The consensus sequence tandemly enhanced transgene expression when it was inserted into the eukaryotic expression vector; the effect of the addition upstream was stronger than that downstream. Thus, we found a MAR consensus sequence that may regulate the MAR-mediated increase in transgene expression.


Subject(s)
Consensus Sequence/genetics , Matrix Attachment Regions/genetics , Transfection , Animals , Base Sequence , CHO Cells , Cricetinae , Cricetulus , Gene Dosage , Gene Expression , Green Fluorescent Proteins/metabolism , Humans , Recombinant Proteins/metabolism , Transgenes
11.
Cancer Sci ; 109(12): 3865-3873, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30259603

ABSTRACT

Thymoquinone (TQ, 2-methyl-5-isopropyl-1,4-benzoquinone), a bioactive constituent extracted from the seeds of Nigella sativa, has been proved to exert anti-tumor efficiency in various cancers. Autophagy is a self-digestion phenomenon, and its role in tumor formation and progression remains controversial. In the present study, we investigated the effects of TQ on renal cell cancer (RCC) cell lines (786-O and ACHN) using wound healing assay, transwell assay and western blot analysis. We found that TQ effectively inhibited the metastatic capacity of RCC cells in vitro, which was also verified in a xenograft model. Meanwhile, we observed LC3 puncta and detected the expression of LC3 in TQ-treated RCC cells, and then found that autophagy was induced by TQ in 786-O and ACHN cell lines. In addition, TQ inhibited the migration and invasion as well as the EMT in RCC cells in an autophagy-dependent manner. To further explore the underlying mechanism, we detected the AMPK/mTOR signaling pathway. The results indicated that TQ inhibited the metastasis of RCC cells by inducing autophagy via AMPK/mTOR signaling pathway. In conclusion, our findings provide a novel therapeutic strategy that aims at TQ-induced autophagy in RCC treatment.


Subject(s)
AMP-Activated Protein Kinases/metabolism , Benzoquinones/administration & dosage , Carcinoma, Renal Cell/drug therapy , Kidney Neoplasms/drug therapy , TOR Serine-Threonine Kinases/metabolism , Animals , Autophagy , Benzoquinones/pharmacology , Carcinoma, Renal Cell/metabolism , Cell Line, Tumor , Cell Movement , Cell Proliferation/drug effects , Cell Survival/drug effects , Humans , Kidney Neoplasms/metabolism , Mice , Neoplasm Metastasis , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
12.
Cancer Sci ; 109(5): 1503-1512, 2018 May.
Article in English | MEDLINE | ID: mdl-29575477

ABSTRACT

Distinguishing the surface markers of cancer stem cells (CSCs) is a useful method for early diagnosis and treatment of tumors, as CSCs may participate in tumorigenesis and metastasis by migrating into the circulatory system. However, the potential targets of CSCs are expressed at low levels in the natural state and are always changing. Thus, dynamic screening has been reported to be an effective measure for exploring CSC markers. In recent years, diverse single-chain variable fragments (scFvs) have been widely used in immunotherapy. In this study, we determined that the scFvs, screened using RD, had a high affinity to microspheres and could inhibit their progression. We also observed that the selected scFvs underwent evolution in vitro, and antitumor-associated proteins were successfully expressed. Combined with chemotherapy, the scFvs had a synergistic effect on the inhibition of the microspheres' progression in vitro and in vivo, which could be ascribed to their high affinity for stem-like cells and the inhibition of the microspheres' collective behaviors. In addition, proteins inhibiting CD44+ /CD24+ and MAPK were involved. Our data indicated that dynamic screening of the scFvs in a natural state was of great significance in the inhibition of the microspheres in vitro and in vivo.


Subject(s)
Breast Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Ribosomes/genetics , Single-Chain Antibodies/pharmacology , Animals , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Surface Display Techniques , Drug Synergism , Drug Therapy , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Mice , Microspheres , Neoplastic Stem Cells/metabolism , Ribosomes/immunology , Xenograft Model Antitumor Assays
13.
Radiology ; 287(1): 137-145, 2018 04.
Article in English | MEDLINE | ID: mdl-29232185

ABSTRACT

Purpose To test the hypothesis that biomarkers of fluorine 18 (18F) fluorodeoxyglucose (FDG) positron emission tomography (PET) can be used for the early detection of therapeutic response to irreversible electroporation (IRE) of liver tumor in a rodent liver tumor model. Materials and Methods The institutional animal care and use committee approved this study. Rats were inoculated with McA-RH7777 liver tumor cells in the left median and left lateral lobes. Tumors were allowed to grow for 7 days to reach a size typically at least 5 mm in longest diameter, as verified with magnetic resonance (MR) imaging. IRE electrodes were inserted, and eight 100-µsec, 2000-V pulses were applied to ablate the tumor tissue in the left median lobe. Tumor in the left lateral lobe served as a control in each animal. PET/computed tomography (CT) and MR imaging measurements were performed at baseline and 3 days after IRE for each animal. Additional MR imaging measurements were obtained 14 days after IRE. After 14-day follow-up MR imaging, rats were euthanized and tumors harvested for hematoxylin-eosin, CD34, and caspase-3 staining. Change in the maximum standardized uptake value (ΔSUVmax) was calculated 3 days after IRE. The maximum lesion diameter change (ΔDmax) was measured 14 days after IRE by using axial T2-weighted imaging. ΔSUVmax and ΔDmax were compared. The apoptosis index was calculated by using caspase-3-stained slices of apoptotic tumor cells. Pearson correlation coefficients were calculated to assess the relationship between ΔSUVmax at 3 days and ΔDmax (or apoptosis index) at 14 days after IRE treatment. Results ΔSUVmax, ΔDmax, and apoptosis index significantly differed between treated and untreated tumors (P < .001 for all). In treated tumors, there was a strong correlation between ΔSUVmax 3 days after IRE and ΔDmax 14 days after IRE (R = 0.66, P = .01) and between ΔSUVmax 3 days after IRE and apoptosis index 14 days after IRE (R = 0.57, P = .04). Conclusion 18F-FDG PET imaging biomarkers can be used for the early detection of therapeutic response to IRE treatment of liver tumors in a rodent model. © RSNA, 2017.


Subject(s)
Electroporation/methods , Fluorodeoxyglucose F18 , Liver Neoplasms/metabolism , Liver Neoplasms/therapy , Positron-Emission Tomography/methods , Radiopharmaceuticals , Animals , Biomarkers/metabolism , Disease Models, Animal , Liver/diagnostic imaging , Liver/metabolism , Liver Neoplasms/diagnostic imaging , Rats , Treatment Outcome
14.
Zhong Nan Da Xue Xue Bao Yi Xue Ban ; 42(2): 184-188, 2017 Feb 28.
Article in Zh | MEDLINE | ID: mdl-28255121

ABSTRACT

OBJECTIVE: To explore the value of ultrasound-guided microwave ablation with artificial pleural effusion for liver tumor adjacent to diaphragmatic dome.
 Methods: A total of 34 patients with liver tumors located at diaphragmatic dome in Hunan Provincial Tumor Hospital were recruited from January 2014 to October 2015. The number of lesions ≤3 or lesion diameter ≤5 cm was in line with the microwave ablation indications. B ultrasound-guided microwave ablation for the liver tumors was undertaken after the artificial pleural effusion being established. 3-4 weeks later after the microwave ablation, all patients were imaged with enhance CT or MRI. The effect of ablation and the complications were evaluated.
 Results: There were 49 lesions in 34 patients, including 30 cases (88.2%) of complete ablation (CA), 3 cases (8.8%) of partial ablation (PA) and one case with new lesions after ablation (2.9%). Thirty-four patients had (1 580±230.7) mL of pleural effusion volume, while one case had bloody pleural effusion. One case had a diaphragmatic thermal injury, and one case had a biliary tumor infection. All of them showed remission after symptomatic treatment. 
 Conclusion: Combination of ultrasound-guided microwave ablation with artificial pleural effusion is a safe and effective therapy for liver tumor adjacent to diaphragmatic dome.


Subject(s)
Ablation Techniques/methods , Isotonic Solutions/administration & dosage , Liver Neoplasms/diagnostic imaging , Liver Neoplasms/therapy , Microwaves/therapeutic use , Diaphragm/diagnostic imaging , Humans , Liver Neoplasms/pathology , Magnetic Resonance Imaging , Tomography, X-Ray Computed , Ultrasonography, Interventional/methods
15.
Int J Cancer ; 139(4): 928-37, 2016 Aug 15.
Article in English | MEDLINE | ID: mdl-27038145

ABSTRACT

The purpose of our study was to test the hypothesis that sorafenib-related dermatologic adverse events (AEs) as an early biomarker can predict the long-term outcomes following the combination therapy of transarterial chemoembolization (TACE) plus sorafenib (TACE-S). The intermediate-stage hepatocellular carcinoma patients who received either TACE-S or TACE-alone treatment were consecutively included into analysis. In the TACE-S group, patients with ≥ grade 2 dermatologic AEs within the first month of sorafenib initiation were defined as responders; whereas those with < grade 2 were defined as nonresponders. In the TACE-S group, the median overall survival (OS) of the responders was significantly longer than that of nonresponders (28.9 months vs. 16.8 months, respectively; p = 0.004). Multivariate analysis demonstrated that nonresponders were significantly associated with an increased risk of death compared with responders (HR = 1.9; 95% confidence Interval-CI: 1.3-2.7; p = 0.001). The survival analysis showed that the median OS was 27.9 months (95% CI: 25.0-30.8) among responders treated with TACE-S vs.18.3 months (95% CI: 14.5-22.1) among those who received TACE-alone (p = 0.046). The median time to progression was 13.1 months (95% CI: 4.4-21.8) in the TACE-S group, a duration that was significantly longer than that in the TACE-alone group [5 months (95% CI: 6.4-13.3), p = 0.014]. This study demonstrated that sorafenib-related dermatologic AEs are clinical biomarkers to identify responders from all of the patients for TACE-S therapy. Sorafenib-related dermatologic AEs, clinical biomarkers, can predict the efficacy of TACE-S in future randomized controlled trials.


Subject(s)
Carcinoma, Hepatocellular/therapy , Chemoembolization, Therapeutic , Liver Neoplasms/therapy , Niacinamide/analogs & derivatives , Phenylurea Compounds/administration & dosage , Adolescent , Adult , Aged , Aged, 80 and over , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/mortality , Chemoembolization, Therapeutic/adverse effects , Chemoembolization, Therapeutic/methods , Disease Progression , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/diagnosis , Liver Neoplasms/etiology , Liver Neoplasms/mortality , Male , Middle Aged , Neoplasm Staging , Niacinamide/administration & dosage , Niacinamide/adverse effects , Phenylurea Compounds/adverse effects , Prognosis , Retrospective Studies , Sorafenib , Treatment Outcome , Young Adult
16.
Mol Cancer ; 14: 28, 2015 Feb 03.
Article in English | MEDLINE | ID: mdl-25645291

ABSTRACT

BACKGROUND: The epithelial-mesenchymal transition (EMT) is crucial for the invasion and metastasis of breast cancer. However, how Notch signaling regulates the EMT process and invasion in breast cancer remains largely unknown. METHODS: The impact of Notch1 silencing by specific shRNAs on the EMT and invasion of human breast cancer MCF-7 and MDA-MB-231 cells as well as xenografts was tested by western blot, real-time polymerase chain reaction (RT-PCR), immunofluorescence, transwell, and immunohistochemistry assays. The effect of Slug silencing or upregulation on the EMT and invasion of breast cancer cells was analyzed, and the effect of Notch1 signaling on Slug expression was determined by the luciferase reporter assay. RESULTS: The Notch1 intracellular domain (N1ICD) and Jagged1 were expressed in breast cancer cells. Notch1 silencing reversed the spontaneous EMT process and inhibited the migration and invasion of breast cancer cells and the growth of xenograft breast cancers. The expression of N1ICD was upregulated significantly by Jagged1-mediated Notch signaling activation. Moreover, Jagged1-mediated Notch signaling promoted the EMT process, migration, and invasion of breast cancer cells, which were abrogated by Notch silencing. Furthermore, the N1ICD positively regulated the Slug expression by inducing Slug promoter activation. Importantly, the knockdown of Slug weakened the invasion ability of breast cancer cells and reversed the Jagged1-induced EMT process with significantly decreased expression of vimentin and increased expression of E-cadherin. In addition, Slug overexpression restored the Notch1 knockdown-suppressed EMT process. CONCLUSIONS: Our novel data indicate that Notch signaling positively regulates the EMT, invasion, and growth of breast cancer cells by inducing Slug expression. The Notch1-Slug signaling axis may represent a potential therapeutic target for breast cancer therapy.


Subject(s)
Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Receptor, Notch1/metabolism , Signal Transduction , Transcription Factors/metabolism , Breast Neoplasms/genetics , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Line, Tumor , Cell Movement/genetics , Epithelial-Mesenchymal Transition/genetics , Female , Gene Expression , Gene Knockdown Techniques , Gene Silencing , Humans , Intercellular Signaling Peptides and Proteins/genetics , Intercellular Signaling Peptides and Proteins/metabolism , Jagged-1 Protein , Membrane Proteins/genetics , Membrane Proteins/metabolism , Neoplasm Invasiveness , Promoter Regions, Genetic , Receptor, Notch1/genetics , Serrate-Jagged Proteins , Snail Family Transcription Factors , Transcription Factors/genetics , Transcriptional Activation
17.
Tumour Biol ; 36(10): 8167-75, 2015 Sep.
Article in English | MEDLINE | ID: mdl-25990455

ABSTRACT

Antimicrobial peptides (AMPs) with Asn-Gly-Arg (NGR) motif have potent cytotoxicity, preferably against tumor cells due to their binding to CD13 on tumor cells. However, the importance of αvß3 expression for antitumor activity of AMPs containing NGR has not been clarified. This study was aimed at designing a new AMP containing NGR and testing their biological activity against different types of tumor cells with varying CD13 and αvß3 expression. We first synthesized the new AMP containing NGR motif (CK21), which effectively entered into CD13+ HT-1080, but less into CD13- αvß3+ MDA-MB-435 and further less into stable αvß3-silencing MDA-MB-435 cells. Furthermore, CK21 displayed dose-dependent antiproliferation against these tumor cells and induced cell cycling arrest at G2/M phases and apoptosis of these tumor cells. In addition, CK21 inhibited the invasion of these tumor cells in vitro and inhibited the growth of implanted tumor cells in vivo. Particularly, the antitumor effect of CK21 in CD13+ HT-1080 was stronger than that of CD13- αvß3+ MDA-MB-435 and much stronger than that of stable αvß3-silencing MDA-MB-435. Our data indicated that the new AMPs containing NGR had potent antitumor activity against CD13+ or αvß3+ tumor cells, preferably against CD13+ tumor cells, possibly through binding to CD13 or αvß3 on tumor cells.


Subject(s)
Anti-Bacterial Agents/pharmacology , CD13 Antigens/metabolism , Cell Proliferation/drug effects , Neoplasms/drug therapy , Neoplasms/pathology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Animals , Apoptosis/drug effects , Blotting, Western , Cell Movement/drug effects , Flow Cytometry , Humans , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Neoplasms/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
18.
Fa Yi Xue Za Zhi ; 31(1): 44-7, 2015 Feb.
Article in Zh | MEDLINE | ID: mdl-26058134

ABSTRACT

Informed consent right is not just for basic ethical consideration, but is important for protecting patient's right by law, which is expressed through informed consent contract. The appraised individuals of forensic clinical examination have the similar legal status as the patients in medical system. However, the law does not require informed consent right for the appraised individuals. I recommend giving certain informed consent right to the appraised individuals in the forensic clinical examination. Under the contracted relationship with the institution, the appraised individuals could participate in the examination process, know the necessary information, and make a selected consent on the examination results, which can assure the justice and fairness of judicial examination procedure.


Subject(s)
Forensic Medicine , Informed Consent , Patient Participation , Humans
19.
Heliyon ; 10(1): e23168, 2024 Jan 15.
Article in English | MEDLINE | ID: mdl-38187319

ABSTRACT

microRNAs (miRNAs) are closely related to the progression of hepatocellular carcinoma (HCC). Cancer-derived exosomes play an essential role in the establishment of the HCC microenvironment. However, the possible effects and underlying mechanisms of exosome (exo) microRNA-23a-5p (miR-23a-5p) in the progression of HCC remain unknown. In this study, we aimed to determine the role and specific molecular mechanism of exo miR-23a-5p in regulating HCC progression and to investigate whether exo miR-23a-5p levels can serve as an indicator of the prognosis of transarterial chemoembolization in patients with HCC. Our findings illustrated that miR-23a-5p was downregulated in exosomes separated from the serum of HCC patients and that miR-23a-5p carried by exosomes inhibited HCC cell proliferation and angiogenesis. Mechanistically, miR-23a-5p negatively targeted peroxiredoxin-2 (PRDX2). Functionally, PRDX2 overexpression relieved exosome-induced inhibition of HCC cell proliferation and angiogenesis by promoting vascular endothelial growth factor (VEGF) expression. In conclusion, Exo miR-23a-5p inhibited HCC proliferation and angiogenesis by regulating PRDX2 expression. Our results revealed the role and specific molecular mechanism of exo miR-23a-5p in regulating HCC progression.

20.
Int J Biol Sci ; 20(2): 446-463, 2024.
Article in English | MEDLINE | ID: mdl-38169627

ABSTRACT

Triple-negative breast cancer (TNBC) has long been considered a major clinical challenge due to its aggressive behavior and poor prognosis. Cancer stem cells (CSCs) are known as the main cells responsible for tumor origination, progression, recurrence and metastasis. Here, we report that M2-type tumor-associated macrophages (TAMs) contribute to cancer stemness in TNBC cells via the secretion of VEGFA. Reciprocally, elevated VEGFA expression by TAM-educated TNBC cells acts as a regulator of macrophage polarization, therefore constitute a feed-back loop between TNBC cells and TAMs. Mechanistically, VEGFA facilitates the CSC phenotype via the NRP-1 receptor and downstream GAPVD1/Wnt/ß-catenin signaling pathway in TNBC cells. Our study underscores the crosstalk between TNBC cells and TAMs mediated by VEGFA and further clarifies the role and underlying mechanisms of the VEGFA/NRP-1/GAPVD1 axis in regulating cancer stemness. We also document an immunosuppressive function of VEGFA in the tumor microenvironment (TME). Therefore, the present study indicates crosstalk between TNBC cells and TAMs induced by VEGFA and provides a potential implication for the combination of immunotherapy and VEGFA-targeted agents in TNBC therapy.


Subject(s)
Antineoplastic Agents , Triple Negative Breast Neoplasms , Humans , Triple Negative Breast Neoplasms/drug therapy , Cell Line, Tumor , Macrophages/metabolism , Antineoplastic Agents/pharmacology , Wnt Signaling Pathway , Tumor Microenvironment/genetics , Vascular Endothelial Growth Factor A/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL