Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 42
1.
J Clin Oncol ; : JCO2302009, 2024 May 06.
Article En | MEDLINE | ID: mdl-38710003

PURPOSE: This phase 3 trial aimed to compare the efficacy and safety of capecitabine or capecitabine plus oxaliplatin (XELOX) with those of fluorouracil plus cisplatin (PF) in definitive concurrent chemoradiotherapy (DCRT) for inoperable locally advanced esophageal squamous cell carcinoma (ESCC). METHODS: Patients were randomly assigned to receive two cycles of capecitabine, XELOX, or PF along with concurrent intensity-modulated radiation therapy. Patients in each arm were again randomly assigned to receive two cycles of consolidation chemotherapy or not. The primary end points were 2-year overall survival (OS) rate and incidence of grade ≥3 adverse events (AEs). RESULTS: A total of 246 patients were randomly assigned into the capecitabine (n = 80), XELOX (n = 85), and PF (n = 81) arms. In capecitabine, XELOX, and PF arms, the 2-year OS rate was 75%, 66.7%, and 70.9% (capecitabine v PF: hazard ratio [HR], 0.91 [95% CI, 0.61 to 1.35]; nominal P = .637; XELOX v PF: 0.86 [95% CI, 0.58 to 1.27]; P = .444); the median OS was 40.9 (95% CI, 34.4 to 49.9), 41.9 (95% CI, 28.6 to 52.1), and 35.4 (95% CI, 30.4 to 45.4) months. The incidence of grade ≥3 AEs during the entire treatment was 28.8%, 36.5%, and 45.7%, respectively. Comparing the consolidation chemotherapy with the nonconsolidation chemotherapy groups, the median OS was 41.9 (95% CI, 34.6 to 52.8) versus 36.9 (95% CI, 28.5 to 44) months (HR, 0.71 [95% CI, 0.52 to 0.99]; nominal P = .0403). CONCLUSION: Capecitabine or XELOX did not significantly improve the 2-year OS rate over PF in DCRT for inoperable locally advanced ESCC. Capecitabine showed a lower incidence of grade ≥3 AEs than PF did.

2.
BMJ ; 383: e076065, 2023 10 31.
Article En | MEDLINE | ID: mdl-37907210

OBJECTIVE: To assess the efficacy and safety of pyrotinib (an irreversible pan-HER (human epidermal growth factor receptor) inhibitor), trastuzumab, and docetaxel compared with placebo, trastuzumab, and docetaxel for untreated HER2 positive metastatic breast cancer. DESIGN: Randomised, double blind, placebo controlled, multicentre, phase 3 trial. SETTING: 40 centres in China between 6 May 2019 and 17 January 2022. PARTICIPANTS: 590 female patients (median age 52 (interquartile range 46-58) years) with untreated HER2 positive metastatic breast cancer. INTERVENTIONS: Eligible patients were randomised 1:1 to receive either oral pyrotinib (400 mg once daily) or placebo, both combined with intravenous trastuzumab (8 mg/kg in cycle 1 and 6 mg/kg in subsequent cycles) and docetaxel (75 mg/m2) on day 1 of each 21 day cycle. Randomisation was stratified by treatment history of trastuzumab in the (neo)adjuvant setting and hormone receptor status. Patients, investigators, and the sponsor's study team were masked to treatment assignment. MAIN OUTCOME MEASURES: The primary endpoint was progression-free survival as assessed by the investigator. RESULTS: Of the 590 randomised patients, 297 received pyrotinib, trastuzumab, and docetaxel treatment (pyrotinib group), and 293 received placebo, trastuzumab, and docetaxel treatment (placebo group). At data cut-off on 25 May 2022, the median follow-up was 15.5 months. The median progression-free survival according to the investigator was significantly longer in the pyrotinib group than in the placebo group (24.3 (95% confidence interval 19.1 to 33.0) months versus 10.4 (9.3 to 12.3) months; hazard ratio 0.41 (95% confidence interval 0.32 to 0.53); one sided P<0.001). Treatment related adverse events of grade 3 or higher were reported in 267 (90%) of the 297 patients in the pyrotinib group and 224 (76%) of the 293 patients in the placebo group. No treatment related deaths occurred in the pyrotinib group, and one (<1%; diabetic hyperosmolar coma) treatment related death occurred in the placebo group. Survival and toxicities are still under assessment with longer follow-up. CONCLUSIONS: Pyrotinib, trastuzumab, and docetaxel showed superiority by significantly improving progression-free survival compared with placebo, trastuzumab, and docetaxel in patients with untreated HER2 positive metastatic breast cancer. The toxicity was manageable. The findings support this dual anti-HER2 regimen as an alternative first line treatment option in this patient population. TRIAL REGISTRATION: ClinicalTrials.gov NCT03863223.


Breast Neoplasms , Female , Humans , Middle Aged , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Docetaxel/adverse effects , Docetaxel/therapeutic use , Double-Blind Method , Receptor, ErbB-2/metabolism , Trastuzumab/adverse effects , Trastuzumab/therapeutic use , Treatment Outcome
3.
Ann Transl Med ; 11(2): 83, 2023 Jan 31.
Article En | MEDLINE | ID: mdl-36819490

Background: Breast cancer is the most common cancer worldwide, and triple-negative breast cancer (TNBC) has the worst prognosis. Standard systemic treatment includes chemotherapy and immunotherapy. Poly ADP-ribose polymerase (PARP) inhibitors are considered in breast cancer (BRCA) susceptibility genes mutated tumors. The role of antiangiogenic drugs is controversial. Immunotherapy with immune checkpoint inhibitor is now a standard of care for TNBC in the US, but its use in combination with anlotinib, an inhibitor of angiogenesis, on TNBC cells was never investigated. Methods: We tested the effects of anlotinib and programmed cell death-ligand 1 (PD-L1) inhibitor on the proliferation, apoptosis, migration, and invasion of MDA-MB-468 and BT-549 TNBC cells through 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide (MTT) assays, cell apoptosis assay, wound healing and transwell matrix assays, and verified whether the combination of the two drugs had synergistic effect. Western blotting was used to detect the effect of anlotinib and PD-L1 inhibitor on the protein expression levels of PI3K, p-PI3K, AKT, p-AKT, Bcl-xl in MDA-MB-468 and BT-549 cells. The effects of anlotinib, PD-L1 inhibitor and the combination of the two drugs on the transplanted tumor of TNBC mice were tested by animal experiments. Results: Anlotinib and PD-L1 inhibitor inhibited the proliferation and promote cell apoptosis of MDA-MB-468 and BT-549 cells, and the combination demonstrated the synergetic effect. Anlotinib and PD-L1 inhibitor inhibited cell migration and invasion, and the effect was strongest in the combination group. Both anlotinib and PD-L1 inhibitor reduced the expression of p-PI3K, p-AKT and Bcl-xl proteins in cells and the effects were the strongest in the combination group. Both anlotinib and PD-L1 inhibitor inhibited the growth of transplanted tumors in mice, and the combined group demonstrated the strongest growth suppression. Conclusions: Anlotinib and PD-L1 inhibitor can inhibit cell proliferation, migration, and invasion of TNBC and promote cell apoptosis, and the two drugs show combined anti-tumor effects in vivo and in vitro. The combination of anlotinib and PD-L1 inhibitor may promote apoptosis of TNBC cells through PI3K/AKT/Bcl-xl signaling pathways, which might offer potential clinical treatment roles for these.

4.
J Hazard Mater ; 443(Pt A): 130206, 2023 02 05.
Article En | MEDLINE | ID: mdl-36279652

Understanding the molecular mechanism of tolerance to heavy metals in hyperaccumulators is important for improving the efficiency of phytoremediation and is interesting for evolutionary studies on plant adaption to abiotic stress. Celosia argentea Linn. was recently discovered to hyperaccumulate both manganese (Mn) and cadmium (Cd). However, the molecular mechanisms underlying Mn and Cd detoxification in C. argentea are poorly understood. Laboratory studies were conducted using C. argentea seedlings exposed to 360 µM Mn and 8.9 µM Cd hydroponic solutions. Plant leaves were analyzed using transcriptional and metabolomic techniques. A total of 3960 differentially expressed genes (DEGs) in plants were identified under Cd stress, among which 17 were associated with metal transport, and 10 belonged to the ATP transporter families. Exposures to Mn or Cd led to the differential expression of three metal transport genes (HMA3, ABCC15, and ATPase 4). In addition, 33 and 77 differentially expressed metabolites (DEMs) were identified under Mn and Cd stresses, respectively. Metabolic pathway analysis showed that the ABC transporter pathway was the most affected in Mn/Cd exposed seedlings. Conjoint transcriptome and metabolome analysis showed that the glutathione (GSH) metabolic pathway was over-represented in the KEGG pathway of both DEGs and DEMs. Our results confirm that the ABC transporter and GSH metabolic pathways play important roles in Mn and Cd detoxification. These findings provide new insight into the molecular mechanisms of tolerance to Mn and Cd toxicity in plants.


Cadmium , Celosia , Cadmium/toxicity , Cadmium/metabolism , Celosia/metabolism , Manganese/toxicity , Manganese/metabolism , Transcriptome , Seedlings/metabolism , Plants/metabolism , Metabolome , ATP-Binding Cassette Transporters/metabolism , Plant Roots/metabolism
5.
BMC Med ; 20(1): 408, 2022 10 25.
Article En | MEDLINE | ID: mdl-36280870

BACKGROUND: Dual inhibition of PD-1/PD-L1 and TGF-ß pathways is a rational therapeutic strategy for malignancies. SHR-1701 is a new bifunctional fusion protein composed of a monoclonal antibody against PD-L1 fused with the extracellular domain of TGF-ß receptor II. This first-in-human trial aimed to assess SHR-1701 in pretreated advanced solid tumors and find the population who could benefit from SHR-1701. METHODS: This was a dose-escalation, dose-expansion, and clinical-expansion phase 1 study. Dose escalation was initiated by accelerated titration (1 mg/kg q3w; intravenous infusion) and then switched to a 3+3 scheme (3, 10, 20, and 30 mg/kg q3w and 30 mg/kg q2w), followed by dose expansion at 10, 20, and 30 mg/kg q3w and 30 mg/kg q2w. The primary endpoints of the dose-escalation and dose-expansion parts were the maximum tolerated dose and recommended phase 2 dose. In the clinical-expansion part, selected tumors were enrolled to receive SHR-1701 at the recommended dose, with a primary endpoint of confirmed objective response rate (ORR). RESULTS: In total, 171 patients were enrolled (dose-escalation: n=17; dose-expansion, n=33; clinical-expansion, n=121). In the dose-escalation part, no dose-limiting toxicity was observed, and the maximum tolerated dose was not reached. SHR-1701 showed a linear dose-exposure relationship and the highest ORR at 30 mg/kg every 3 weeks, without obviously aggravated toxicities across doses in the dose-escalation and dose-expansion parts. Combined, 30 mg/kg every 3 weeks was determined as the recommended phase 2 dose. In the clinical-expansion part, SHR-1701 showed the most favorable efficacy in the gastric cancer cohort, with an ORR of 20.0% (7/35; 95% CI, 8.4-36.9) and a 12-month overall survival rate of 54.5% (95% CI, 29.5-73.9). Grade ≥3 treatment-related adverse events occurred in 37 of 171 patients (22%), mainly including increased gamma-glutamyltransferase (4%), increased aspartate aminotransferase (3%), anemia (3%), hyponatremia (3%), and rash (2%). Generally, patients with PD-L1 CPS ≥1 or pSMAD2 histochemical score ≥235 had numerically higher ORR. CONCLUSIONS: SHR-1701 showed an acceptable safety profile and encouraging antitumor activity in pretreated advanced solid tumors, especially in gastric cancer, establishing the foundation for further exploration. TRIAL REGISTRATION: ClinicalTrials.gov , NCT03710265.


Stomach Neoplasms , Humans , gamma-Glutamyltransferase/therapeutic use , Programmed Cell Death 1 Receptor , Antibodies, Monoclonal/therapeutic use , Aspartate Aminotransferases/therapeutic use , Transforming Growth Factor beta/therapeutic use , Receptors, Transforming Growth Factor beta/therapeutic use
6.
Front Oncol ; 12: 819714, 2022.
Article En | MEDLINE | ID: mdl-35251986

Poly (ADP-ribose) polymerase inhibitor (PARPi) resistance is a new challenge for antitumor therapy. The purpose of this study was to investigate the reversal effects of chidamide on fluzoparib resistance, a PARPi, and its mechanism of action. A fluzoparib-resistant triple-negative breast cancer (TNBC) cell line was constructed, and the effects of chidamide and fluzoparib on drug-resistant cells were studied in vitro and in vivo. The effects of these drugs on cell proliferation, migration, invasiveness, the cell cycle, and apoptosis were detected using an MTT assay, wound-healing and transwell invasion assays, and flow cytometry. Bioinformatics was used to identify hub drug resistance genes and Western blots were used to assess the expression of PARP, RAD51, MRE11, cleaved Caspase9, and P-CDK1. Xenograft models were established to analyze the effects of these drugs on nude mice. In vivo results showed that chidamide combined with fluzoparib significantly inhibited the proliferation, migration, and invasiveness of drug-resistant cells and restored fluzoparib sensitivity to drug-resistant cells. The combination of chidamide and fluzoparib significantly inhibited the expression of the hub drug resistance genes RAD51 and MRE11, arrested the cell cycle at the G2/M phase, and induced cell apoptosis. The findings of this work show that chidamide combined with fluzoparib has good antineoplastic activity and reverses TNBC cell resistance to fluzoparil by reducing the expression levels of RAD51 and MRE11.

7.
Pest Manag Sci ; 78(3): 1018-1028, 2022 Mar.
Article En | MEDLINE | ID: mdl-34773351

BACKGROUND: Bactrocera dorsalis is a devastating pest on fruits and vegetables because the adult female is the key factor that determines the population density of offspring and the degree of host damage. Unfortunately, there is still a lack of effective female attractants for behavioral control. Males of B. dorsalis fed on methyl eugenol (ME) were shown to be more sexually attracted to females and, therefore, were more successful in mating over ME-deprived males. RESULTS: In the current study, we demonstrated that (E)-coniferyl alcohol (E-CF), one of the ME metabolites in males, was highly attractive to sexually-mature females in laboratory bioassays. During the dusk courtship period, mature females showed the highest response to E-CF. However, there were no significant differences in olfactory responses to E-CF between virgin and mated mature females. Moreover, no obvious signs and symptoms of toxicity or death were observed in mice during a 14-day acute oral toxicity test. Toxicologically, no significant changes were observed in body weight, water intake, food consumption and absolute and relative organ weights between control and treated groups of healthy-looking mice, implying that E-CF could be regarded as non-toxic. Furthermore, cytotoxicity assessment revealed that E-CF was non-toxic against human fetal lung fibroblast 1 (HFL1), human breast cancer (MDA-MB-231), mouse embryonic hepatocytes (BNL-CL.2) and Spodoptera frugiperda ovary (SF-9) cell lines. CONCLUSIONS: E-CF proved to be an effective, promising and eco-friendly lure to B. dorsalis females. Therefore, this study may facilitate the development of novel control strategies against B. dorsalis in the field.


Tephritidae , Animals , Drosophila , Female , Male , Mice , Phenols , Reproduction
8.
BMC Cancer ; 21(1): 1188, 2021 Nov 07.
Article En | MEDLINE | ID: mdl-34743685

BACKGROUND: In recent years, gene expression-based analysis has been used for disease biomarker discovery, providing ways for better diagnosis, leading to improvement of clinical treatment efficacy. This study aimed to explore the role of miR-16-5p and ANLN in breast cancer (BC). METHODS: Cohort datasets of BC were obtained from the Gene Expression Omnibus (GEO) and the Cancer Genome Atlas (TCGA) and analyzed by bioinformatics tools. qRT-PCR and western blotting were applied to validate ANLN and its protein expression. A dual-luciferase reporter assay was used to prove the regulatory relationship of miR-16-5p and ANLN. Finally, MTT, wound healing, Transwell invasion and flow cytometry analyses of the cell cycle and apoptosis were performed to assess cell proliferation, migration, invasion, cell cycle and apoptosis, respectively. RESULTS: A total of 195 differentially expressed genes (DEGs) and 50 overlapping microRNAs (miRNAs) were identified. Among these DEGs and miRNAs, ANLN, associated with poor overall survival in BC, overlapped in the GSE29431, GSE42568, TCGA and GEPIA2 databases. Moreover, ANLN was highly expressed, while miR-16-5p was lower in BC cells than in breast epithelial cells. Then, we confirmed that ANLN was directly targeted by miR-16-5p in BC cells. Over-expression of miR-16-5p and knock-down of ANLN remarkably inhibited cell proliferation and migration as well as cell invasion, arrested the cells in G2/M phase and induced apoptosis in BC cells. CONCLUSIONS: These findings suggest that miR-16-5p restrains proliferation, migration and invasion while affecting cell cycle and promotes apoptosis by regulating ANLN, thereby providing novel candidate biomarkers for the diagnosis and treatment of BC.


Breast Neoplasms/metabolism , Cell Proliferation , MicroRNAs/metabolism , Microfilament Proteins/metabolism , Apoptosis/genetics , Breast/metabolism , Breast/pathology , Breast Neoplasms/genetics , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation/genetics , Databases, Genetic , Female , G2 Phase Cell Cycle Checkpoints , Gene Expression , Humans , M Phase Cell Cycle Checkpoints , Microfilament Proteins/genetics , Neoplasm Invasiveness/genetics , Prognosis , Up-Regulation
9.
Ecotoxicol Environ Saf ; 223: 112567, 2021 Oct 15.
Article En | MEDLINE | ID: mdl-34364125

Males of the Oriental fruit fly Bactrocera dorsalis (Hendel) are highly attracted to, and compulsively feed, on methyl eugenol (ME). ME is converted into 2-allyl-4,5-dimethoxyphenol (DMP) and (E)-coniferyl alcohol (E-CF), which are temporarily sequestered in the fly's rectal gland prior to being released at dusk. Previous research initially confirmed that DMP is a relatively strong lure to B. dorsalis males. However, the characteristics of males' response to DMP and toxicology of DMP remains largely unclear. In our study, we demonstrated that DMP was more attractive to sexually mature males than E-CF tested in laboratory bioassays. Interestingly, the responsiveness of mature males to DMP was not uniform throughout the day, eliciting the highest response during the day and dropping to a low level at night. Furthermore, there were no significant differences between the olfactory responses of virgin and mated mature males to DMP. No obvious signs of toxic symptom and deaths were observed in mice during a 14-day acute oral toxicity testing. Further, toxicologically significant changes were not observed in body weight, water intake, food consumption, and absolute and relative organ weights between control and treated groups, implying DMP could be regarded as nontoxic. Lastly, the cytotoxicity data of DMP on cells showed that it exhibited no significant cytotoxicity to normal human and mouse cells. Taken together, results from both the acute and cellular toxicity experiments demonstrated the nontoxic nature of DMP. In conclusion, DMP shows promise as an effective and eco-friendly lure for B. dorsalis males, and may contribute to controlling B. dorsalis in the flied.


Sex Attractants , Tephritidae , Animals , Eugenol/analogs & derivatives , Male , Mice , Reproduction
10.
Ann Transl Med ; 9(12): 1001, 2021 Jun.
Article En | MEDLINE | ID: mdl-34277801

BACKGROUND: Apatinib is a new generation of small molecule tyrosine kinase inhibitor, which can highly selectively inhibit phosphorylation of vascular endothelial growth factor receptor 2 (VEGFR-2). This study aimed to investigate the synergistic effects of apatinib and paclitaxel (PTX) on triple-negative breast cancer (TNBC) in vivo and in vitro, and to explore the molecular mechanism of the PI3K/p65/Bcl-xl pathway. METHODS: In vitro, 3-(4,5-dimethylthiazol-2-Yl)-2,5-diphenyltetrazolium bromide (MTT) method, flow cytometry (FCM), wound healing assay, and transwell matrix assay were conducted to measure the effects of apatinib and PTX on cell viability, apoptosis, migration, and invasion in TNBC cell line MDA-MB-468. Western blot (WB) was conducted to detect protein expression levels of PI3K, p65, and Bcl-xl after the application of apatinib and PTX. In vivo, MDA-MB-468 tumor-bearing nude mice were treated with apatinib and PTX, and tumor growth was observed. RESULTS: In vitro, apatinib and PTX could synergistically suppress the cell viability, the combined group had the most obvious effect. Apatinib and PTX could promote apoptosis and suppress migration and invasion of TNBC cells. Apatinib could reduce the expression of p-PI3K, p65, and Bcl-xl proteins (P<0.05). In vivo, apatinib and PTX could inhibit tumor size and weight of model mice, and the combined agents had a more significant effect. CONCLUSIONS: Apatinib could enhance the anti-tumor effect of PTX on TNBC cells through the PI3K/p65/Bcl-xl molecular pathway, and apatinib combined with PTX might be a promising option for TNBC treatment.

11.
Front Oncol ; 11: 616443, 2021.
Article En | MEDLINE | ID: mdl-34094901

Pyrotinib (PYR) is a pan-HER kinase inhibitor that inhibits signaling via the RAS/RAF/MEK/MAPK and PI3K/AKT pathways. In this study, we aimed to investigate the antitumor efficacy of pyrotinib combined with adriamycin (ADM) and explore its mechanisms on HER2+ breast cancer. We investigated the effects of PYR and ADM on breast cancer in vitro and in vivo. MTT assay, Wound-healing, and transwell invasion assays were used to determine the effects of PYR, ADM or PYR combined with ADM on cell proliferation, migration, and invasion of SK-BR-3 and AU565 cells in vitro. Cell apoptosis and cycle were detected through flow cytometry. In vivo, xenograft models were established to test the effect of PYR, ADM, or the combined therapy on the nude mice. Western blotting was performed to assess the expression of Akt, p-Akt, p-65, p-p65, and FOXC1. The results indicated that PYR and ADM significantly inhibited the proliferation, migration, and invasion of SK-BR-3 and AU565 cells, and the inhibitory rate of the combination group was higher than each monotherapy group. PYR induced G1 phase cell-cycle arrest, while ADM induced G2 phase arrest, while the combination group induced G2 phase arrest. The combined treatment showed synergistic anticancer activities. Moreover, PYR significantly downregulated the expression of p-Akt, p-p65, and FOXC1. In clinical settings, PYR also exerts satisfactory efficacy against breast cancer. These findings suggest that the combination of PYR and ADM shows synergistic effects both in vitro and in vivo. PYR suppresses the proliferation, migration, and invasion of breast cancers through down-regulation of the Akt/p65/FOXC1 pathway.

12.
Gland Surg ; 10(2): 799-806, 2021 Feb.
Article En | MEDLINE | ID: mdl-33708561

BACKGROUND: Triple negative breast cancer (TNBC) is usually aggressive and accompanied by a poor prognosis. The molecular biological mechanism of TNBC pathogenesis is still unclear, and requires more detailed research. The aim of this study was to screen and verify potential biomarkers of TNBC, and provide new clues for the treatment and diagnosis of TNBC. METHODS: In this work, GSE76250 was downloaded from the Gene Expression Omnibus (GEO) database and included 165 TNBC samples and 33 paired normal breast tissues. The R software and its related software package were used for data processing and analysis. Compared with normal tissues, genes with a false discovery rate (FDR) <0.01 and log fold change (logFC) ≥1 or ≤-1 were identified as differentially expressed genes (DEGs) by limma package. Survival prognoses were analyzed by Kaplan-Meier plotter database. RESULTS: In total, 160 up-regulated and 180 down-regulated genes were identified. The biological mechanism of enrichment analysis presented that DEGs were significantly enriched in chromosome segregation, extracellular matrix, and extracellular matrix structural constituent, among others. A total of 8 hub genes (CCNB1, CDK1, TOP2A, MKI67, TTK, CCNA2, BUB1, and PLK1) were identified by the protein-protein interaction network (PPIN) and Cytoscape software. Survival prognosis of these hub genes showed that they were negatively correlated with overall survival. CONCLUSIONS: The 8 hub genes and pathways that were identified might be involved in tumorigenesis and become new candidate biomarkers for TNBC treatment.

13.
Transl Cancer Res ; 10(5): 2399-2408, 2021 May.
Article En | MEDLINE | ID: mdl-35116555

BACKGROUND: Despite recent advances in screening, treatment, and survival, breast cancer remains the most invasive cancer in women. The development of novel diagnostic and therapeutic markers for breast cancer may provide more information about its pathogenesis and progression. METHODS: We obtained GSE86374 micro-expression matrix chip data from the Gene Expression Omnibus (GEO) database consisting of 159 samples (124 normal samples and 35 breast cancer samples). The language was then used to perform data processing and differential expression analysis. For all differentially expressed genes (DEGs), "FDR <0.01 and |logFC| ≥1" were selected as thresholds. RESULTS: In this study, 173 up-regulated genes and 143 down-regulated genes were selected for GO and KEGG enrichment analysis. These genes are also significantly enriched in the KEGG pathway, including phenylalanine metabolism, staphylococcus aureus infection, and the PPAR signaling pathway. The survival and prognosis of the selected eight key genes (DLGAP5, PRC1, TOP2A, CENPF, RACGAP1, RRM2, PLK1, and ASPM) were analyzed by the Kaplan-Meier plotter database. CONCLUSIONS: Eight hub genes and pathways closely related to the onset and progression of breast cancer were identified. We found that the PPAR signaling pathway, especially PPARγ, plays an important role in breast cancer and suggest this pathway be the subject of further research.

14.
Front Cell Dev Biol ; 9: 785796, 2021.
Article En | MEDLINE | ID: mdl-34977029

HER2+/HR+ breast cancer is a special molecular type of breast cancer. Existing treatment methods are prone to resistance; "precision treatment" is necessary. Pyrotinib is a pan-her kinase inhibitor that can be used in HER2-positive tumors, while SHR6390 is a CDK4/6 inhibitor that can inhibit ER+ breast cancer cell cycle progression and cancer cell proliferation. In cancer cells, HER2 and CDK4/6 signaling pathways could be nonredundant; co-inhibition of both pathways by combination of SHR6390 and pyrotinib may have synergistic anticancer activity on HER2+/HR+ breast cancer. In this study, we determined the synergy of the two-drug combination and underlying molecular mechanisms. We showed that the combination of SHR6390 and pyrotinib synergistically inhibited the proliferation, migration, and invasion of HER2+/HR+ breast cancer cells in vitro. The combination of two drugs induced G1/S phase arrest and apoptosis in HER2+/HR+ breast cancer cell lines. The combination of two drugs prolonged the time to tumor recurrence in the xenograft model system. By second-generation RNA sequencing technology and enrichment analysis of the pyrotinib-resistant cell line, we found that FOXM1 was associated with induced resistance to HER2-targeted therapy. In HER2+/HR+ breast cancer cell lines, the combination of the two drugs could further reduce FOXM1 phosphorylation, thereby enhancing the antitumor effect to a certain extent. These findings suggest that SHR6390 combination with pyrotinib suppresses the proliferation, migration, and invasion of HER2+/HR+ breast cancers through regulation of FOXM1.

15.
Technol Cancer Res Treat ; 19: 1533033820967454, 2020.
Article En | MEDLINE | ID: mdl-33084525

OBJECTIVE: We aimed to evaluate immune-related adverse events occurring in clinical trials of anti-programmed cell death 1 (PD-1) drugs, compared with control treatments, including chemotherapy, targeted drugs, or placebo. Further we compared the occurrence of immune -related events in patients treated with different anti-PD-1 drugs. DATA SOURCES: Randomized controlled trial (RCT) data were sourced from PubMed, Embase, and the Cochrane Central Register of Controlled Trials combined with https://clinicaltrials.gov. METHODS: Randomized controlled trial of anti-PD-1 drugs compared with control treatments published between January 1, 1970 and March 1,2019, were searched and data on trial patient characteristics, and adverse events extracted, reviewed, and subjected to meta-analysis. RESULTS: Eighteen Randomized controlled trials were included in our study. The Randomized controlled trials compared nivolumab (n = 12), pembrolizumab (n = 6), with chemotherapy (n = 13), targeted drugs (n = 2), or placebo (n = 3). Compared with the control group, the risk of any immune-related adverse events in patients treated with anti-PD-1 drugs was increased (RR, 2.65; 95% confidence interval, 1.84-3.83; P < 0.00001). Of the immune-related adverse events, the risk rates of pneumonitis (risk ratio, 2.10; 95% CI, 0.85-5.18), colitis (2.96;1.62-5.38), hypophysitis(4.79;1.54-14.89), hypothyroidism(7.87;5.36-11.57), hyperthyroidism (7.03;4.35-11.34), rash (1.58;0.98-2.54), pruritus (2.28; 1.38-3.76), and hepatitis (9.31;2.18-39.85) were increased by anti-PD-1 drugs. Further, the risk of immune-related adverse events was similar for patients treated with pembrolizumab and nivolumab (P = 0.14). CONCLUSIONS: In addition to previously reported organ-specific immune-related adverse events, we found that the risk of hyperthyroidism was also increased, in anit-PD-1-treated patients, relative to control treatments. The risk of total immune-related adverse events, was similar for pembrolizumab and nivolumab.


Antibodies, Monoclonal, Humanized/therapeutic use , Drug-Related Side Effects and Adverse Reactions/epidemiology , Immune Checkpoint Inhibitors/therapeutic use , Programmed Cell Death 1 Receptor/genetics , Antibodies, Monoclonal, Humanized/adverse effects , Drug-Related Side Effects and Adverse Reactions/pathology , Humans , Immune Checkpoint Inhibitors/adverse effects , Nivolumab/adverse effects , Nivolumab/therapeutic use , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/immunology , Randomized Controlled Trials as Topic
16.
Technol Cancer Res Treat ; 19: 1533033820960721, 2020.
Article En | MEDLINE | ID: mdl-32990165

BACKGROUND: To investigate the pathologic complete response (pCR) rates of dual human epidermal growth factor receptor 2 (HER2) blockade in a neoadjuvant setting for HER2+ breast cancer. METHODS: We searched randomized clinical trials (RCTs) using dual HER2 blockade in a neoadjuvant setting for HER2+ breast cancer in PubMed, the Cochrane Library, Embase and ClinicalTrials.gov up to July 5, 2020, and all included studies were assessed according to the Cochrane Collaboration tool for assessing the risk of bias of RCTs, and the statistical analyses were performed using STATA 14.0 software. RESULTS: A total of 9 RCTs involving 2758 patients were included. Meta-analysis indicated that the pCR rates of lapatinib/pertuzumab/neratinib plus trastuzumab versus trastuzumab [relative risk (RR) = 1.31; 95% confidence interval (CI): 1.21-1.43; p < 0.001)] and lapatinib plus trastuzumab versus lapatinib (RR = 1.39; 95%CI: 1.25-1.53; p < 0.001) showed a significant statistical difference between dual HER2-blockade treatment and single-agent treatment in a neoadjuvant setting for HER2+ breast cancer. Additionally, there was no statistically significant difference in disease-free survival (HR = 0.72; 95% CI: 0.47-1.09; p = 0.123), incidence of serious adverse events (SAEs) (RR = 1.04; 95%CI: 0.81-1.33; p = 0.778) and cardiotoxicity(RR = 1.30; 95%CI: 0.81-2.08; p = 0.280), and the pCR rate was unaffected by hormone receptor status. CONCLUSIONS: The pCR rate of neoadjuvant dual-target therapy for HER2+ breast cancer was significantly higher than that of single-target therapy. Furthermore, the results indicated that the safety of dual-target therapy is similar to that of single-target therapy.


Antineoplastic Agents, Immunological/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/drug therapy , Neoadjuvant Therapy/methods , Receptor, ErbB-2/antagonists & inhibitors , Antibodies, Monoclonal, Humanized/therapeutic use , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Disease-Free Survival , Female , Humans , Lapatinib/therapeutic use , Molecular Targeted Therapy/methods , Quinolines/therapeutic use , Receptor, ErbB-2/metabolism , Trastuzumab/therapeutic use
17.
Environ Pollut ; 266(Pt 1): 115408, 2020 Nov.
Article En | MEDLINE | ID: mdl-32829173

Phytoextraction using Celosia argentea Linn. can potentially decontaminate Cd-contaminated soils. However, most earlier studies have been conducted at laboratory scale and for a relatively short remediation period. To evaluate the phytoextraction efficiency of C. argentea combined with different soil amendments (ammonium chloride, Bacillus megaterium, and citric acid), an 18-month field experiment was carried out in a farmland soil contaminated with 3.68 mg kg-1 Cd by mine tailings in southern China. Soil Cd concentrations were decreased by 6.34 ± 0.73% after the three harvestings (with no amendments), which was 2.27 times that of the no-planting control (p < 0.05). Application of ammonium chloride, B. megaterium, and citric acid increased the overall Cd reduction rate in soil by 40.5%, 46.1%, and 105%, respectively. The application of citric acid decreased total Cd in soil by up to 16.9% in the rhizosphere soil and 13.0% in the bulk soil. The highest annual shoot biomass yield and Cd extraction amount reached 8.79 t ha-1 and 273 g ha-1. Acid-soluble Cd fraction in the rhizosphere was significantly lower compared to that in the bulk soil (p < 0.05), which indicates that mobile Cd in the rhizosphere was taken up by the roots vastly. C. argentea phytoextraction also improved soil metabolic functions by increasing the activities of soil enzymes (urease, invertase, phosphatase, and catalase). These findings demonstrate that Cd phytoextraction using C. argentea with the application of soil amendments can greatly improve the quality of Cd-contaminated soils.


Celosia , Soil Pollutants/analysis , Biodegradation, Environmental , Cadmium/analysis , China , Soil
18.
Clin Lab ; 66(4)2020 Apr 01.
Article En | MEDLINE | ID: mdl-32255296

BACKGROUND: This study sought to systematically assess the diagnostic and prognostic value of serum amyloid A (SAA) in gastric cancer (GC). METHODS: PubMed, Embase, EBSCO, CNKI, and the Cochrane Library databases were searched for eligible studies. Extracted data were analyzed to determine diagnostic parameters and the summary receiver operating characteristic (SROC). Pooled hazard ratios (HR) and odds ratios (ORs) with their corresponding 95% confidence intervals (95% CIs) were calculated to summarize the effects. RESULTS: Six articles in English that met the inclusion criteria were identified. Meta-analysis of the included studies indicated high sensitivity of 0.84 (95% CI: 0.77 - 0.89) and moderate specificity of 0.61 (95% CI: 0.55 - 0.67) of SAA, with a diagnostic odds ratio (DOR) of 8.17 (95% CI: 4.82 - 13.86). The area under the receiver operating characteristic curve was 0.77. Survival analysis showed that SAA was associated with shorter survival time (HR = 4.42, p = 0.000; I2 = 0.0%). Stratified analyses showed that the assay of SAA from serum harbored higher efficacy than that from serum + plasma (AUC: 0.81 vs. 0.77), and SAA testing also achieved a better diagnostic performance in Asians than in Caucasians (AUC: 0.77 vs. 0.50). CONCLUSIONS: Collectively, our analyses suggest that SAA can be used as a clinical auxiliary reference index for the diagnosis and prognosis prediction of GC; however, this diagnostic method is not independently sufficient. Our findings require confirmation in a larger prospective study.


Biomarkers, Tumor/blood , Serum Amyloid A Protein/analysis , Stomach Neoplasms/blood , Humans , Prognosis , Sensitivity and Specificity , Stomach Neoplasms/diagnosis , Survival Analysis
19.
J Ovarian Res ; 12(1): 24, 2019 Mar 21.
Article En | MEDLINE | ID: mdl-30898156

OBJECT: Ovarian cancer is the primary cause of cancer-associated deaths among gynaecological malignancies. Increasing evidence suggests that microRNAs may be potential biomarkers for the diagnosis and prognosis of cancer. In this study, we conducted a systematic review and meta-analysis to summarize the global research and to evaluate the overall diagnostic accuracy of miRNAs in detecting ovarian cancer. METHODS: A systematic literature search was conducted for relevant studies through July 20, 2017, in English databases (CENTRAL, MEDLINE, and EMBASE), the Grey reference database and Chinese databases. Statistical analysis was conducted using OpenMetaAnalyst, STATA 14.0 and RevMan 5.3. Pooled sensitivity, specificity, and other parameters were used to assess the overall miRNA assay performance using a bivariate random-effects model (BRM). Meta-regression and subgroup analyses were performed to dissect the heterogeneity. Sensitivity analysis was performed to assess the robustness of our analysis, and the publication bias of the selected studies was assessed using Deeks' funnel plot asymmetry test. RESULTS: Thirteen articles described 33 studies, including 1081 patients with ovarian cancer and 518 controls. The pooled results were as follows: sensitivity, 0.89 (95% CI: 0.84-0.93); specificity, 0.64 (95% CI: 0.56-0.72); positive likelihood ratio, 2.18 (95% CI: 1.89-2.51); negative likelihood ratio, 0.15 (95% CI: 0.11-0.22); and diagnostic odds ratio (DOR), 13.21 (95% CI: 9.00-19.38). We conducted subgroup analyses based on ethnicity, research design, and miRNA profiling and found that multiple miRNA panels were more accurate in detecting ovarian cancer, with a combined DOR of 30.06 (95% CI: 8.58-105.37). CONCLUSION: Per the meta-analysis, circulating miRNAs may be novel and non-invasive biomarkers for detecting ovarian cancer, particularly multiple miRNA panels, which have potential diagnostic value as screening tools in clinical practice.


Biomarkers, Tumor/metabolism , Circulating MicroRNA/metabolism , Ovarian Neoplasms/diagnosis , Female , Humans , Odds Ratio , Ovarian Neoplasms/metabolism , Prognosis , Sensitivity and Specificity
20.
Am J Transl Res ; 10(7): 1935-1948, 2018.
Article En | MEDLINE | ID: mdl-30093933

OBJECTIVE: To assess the diagnostic performance of cell-free DNA assays in the detection of bladder cancer. PATIENTS AND METHODS: The quality of the studies included in this meta-analysis was assessed using the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool. Statistical analyses were performed using the software RevMan 5.3 and Stata 14.0. We assessed the pooled sensitivity and specificity, positive/negative likelihood ratios (PLRs/NLRs), diagnostic odds ratios (DORs), and corresponding 95% confidence intervals (95% CIs). Summary receiver operating characteristic curve (ROC curve) and area under the curve (AUC) were used to summarize the overall test performance. Heterogeneity and publication bias were also examined. RESULTS: Eleven studies included 802 bladder cancer patients and 668 controls met the eligibility criteria. The overall diagnostic accuracy was measured as follows: sensitivity 0.71 (95% CI = 0.64-0.77), specificity 0.78 (95% CI = 0.70-0.85), PLR 3.3 (95% CI = 2.4-54.5), NLR 0.37 (95% CI = 0.30-0.46), DOR 9 (95% CI = 6-14), and AUC 0.80 (95% CI = 0.77-0.83). Subgroup analysis suggested that ethnicity significantly accounted for the heterogeneity of specificity. The Deeks' funnel plot asymmetry test (P = 0.97) suggested no potential publication bias. CONCLUSIONS: Cell-free DNA has a high diagnostic value in bladder cancer.

...