Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
World J Clin Cases ; 11(4): 866-873, 2023 Feb 06.
Article in English | MEDLINE | ID: mdl-36818625

ABSTRACT

BACKGROUND: The advent of molecular targeted agents and immune checkpoint inhibitors has greatly improved the treatment of advanced renal cell carcinoma (RCC), thus significantly improving patient survival. The incidence of rare drug-related adverse events has gained increased attention. CASE SUMMARY: We report a patient with advanced RCC treated with multiple lines of molecular targeted agents and immune checkpoint inhibitors, who developed a pulmonary infection after treatment with everolimus in combination with lenvatinib. Determining the pathogenic organism was difficult, but it was eventually identified as Pneumocystis jirovecii by next-generation sequencing (NGS) of bronchoscopic alveolar lavage fluid (BALF) and successfully treated with trimethoprim-sulfamethoxazole. CONCLUSION: Rare pulmonary infections caused by molecular targeted agents are not uncommon in clinical practice, but their diagnosis is difficult. Evaluating BALF with NGS is a good method for rapid diagnosis of such infections.

2.
Front Oncol ; 12: 966818, 2022.
Article in English | MEDLINE | ID: mdl-36465390

ABSTRACT

Perivascular epithelioid cell tumors (PEComas) are rare mesenchymal tumors arising from perivascular epithelial cells. There was no standard treatment for unresectable PEComa before 2021. For a low incidence and a rarely curable disease, development of new therapy is essential. A 45-year-old female was diagnosed with malignant renal PEComa (likely with TFE3 rearrangement) that underwent rapid progression after 10 months of surgery. The patient then received the tyrosine kinase inhibitor (TKI) Apatinib, and the tumor remained stable for 15 months before another progression. The patient then received the MTOR inhibitor everolimus that alleviated her symptoms but the tumor went into remission again after another 15 months. This result suggests that antagonizing the vascular endothelial growth factor receptor (VEGFR) pathway be a useful strategy for malignant PEComas, along with the MTOR pathway inhibition that had recently been approved for the rare tumor.

3.
J Cancer Res Ther ; 17(5): 1234-1240, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34850772

ABSTRACT

AIMS: Our study assessed the efficacy and safety of sintilimab-based regimens for real-world treatment of advanced gastric and gastroesophageal junction adenocarcinoma (G/GEJAC). MATERIALS AND METHODS: Cases of advanced nonresectable G/GEJAC treated with sintilimab-based regimens in the Department of Gastroenterology of Shanxi Provincial Cancer Hospital between December 2018 and September 2020 were retrospectively examined. Endpoints included median progression-free survival (mPFS), median overall survival (mOS), disease control rate (DCR), objective response rate (ORR), and adverse events (AEs). Univariate and multivariate analyses were conducted to determine the effect of stratification factors on efficacy. RESULTS: Among the 37 included patients, mPFS and mOS were 4.27 and 7.3 months, respectively. Efficacy was evaluated at least once in 32 of 37 patients. The ORR and DCR were 12.5% and 65.63%, respectively. Among four patients with mismatch repair deficiency/microsatellite instability-high (dMMR/MSI-H) lesions, two achieved partial remission, and two displayed stable disease, resulting in a DCR of 100%. The most observed AEs included leukopenia, neutropenia, thrombocytopenia, nausea, and skin rash. mPFS was 4.90 months in patients who received sintilimab in the first- or second-line setting, versus 3.00 months in other patients. A significant survival difference was found between these groups in univariate and multivariate analyses. CONCLUSIONS: The application of sintilimab-based regimens achieved good disease control and tolerability for the real-world treatment of advanced G/GEJAC. The treatment was more effective when administered in the first- or second-line setting. Patients with dMMR/MSI-H lesions may also benefit from sintilimab-based regimens.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal, Humanized/therapeutic use , Esophageal Neoplasms/drug therapy , Esophagogastric Junction/drug effects , Stomach Neoplasms/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Esophageal Neoplasms/pathology , Esophagogastric Junction/pathology , Female , Follow-Up Studies , Humans , Male , Middle Aged , Prognosis , Retrospective Studies , Stomach Neoplasms/pathology , Survival Rate , Young Adult
4.
Article in English | MEDLINE | ID: mdl-34698510

ABSTRACT

Objective: To investigate the effects of M2 macrophage-derived exosomes (M2-Exos) on proliferation, migration, and apoptosis of gastric cancer cells in the tumor microenvironment and to further explore their possible molecular mechanism. Materials and Methods: M2 macrophages were induced from THP-1 cells and identified by qRT-PCR. Exosomes were extracted by ultracentrifugation and identified by transmission electron microscopy (TEM), nanoparticle tracking analysis (NTA), and Western blot analysis. Fluorescence labeling was used to detect the internalization of exosomes in receptors. The proliferation, migration, and invasion of AGS and HGC27 cells were determined by EdU and MTS, wound healing and Transwell assay, and flow cytometry, respectively. Proteins in the related pathway of M2-Exos affecting the progression of gastric cancer were detected by Western blot analysis. Results: In this study, M2 macrophages and M2-Exos were successfully obtained. The purified M2-Exos were observed as small round vesicles with diameters of 50-90 nm and positive expression of CD63, CD9, and TSG101. Besides, M2-Exos can be effectively taken up and internalized by AGS and HGC27 cells. Cell behavior studies showed that M2-Exos promoted proliferation and migration and inhibited the apoptosis of AGS and HGC27. Further research illustrated that M2-Exos promoted the phosphorylation of P38 and high expression of programmed death ligand 1 (PD-L1). Conclusions: This study demonstrated that M2-Exos promoted proliferation and migration and inhibited the apoptosis of gastric cancer cells. Mechanically, M2-Exos may promote gastric cancer progression through the P38MAPK signaling pathway and achieve immune escape through elevating the expression of PD-L1.

5.
Biosci Rep ; 40(2)2020 02 28.
Article in English | MEDLINE | ID: mdl-32022229

ABSTRACT

Radiation therapy is a widely used treatment for esophageal cancer. However, radiation resistance might result in a poor prognosis. Overexpression of HER2 has been related to adaptive radiation resistance. Pyrotinib is a HER2 inhibitor that shows an anti-tumor effect in breast cancer. The present study aims to explore the influence of pyrotinib combined with radiotherapy on HER2-positive esophageal cancer cells and explore the underlying mechanism. We screened two cell lines (TE-1 and KYSE30) that highly express HER2 from several human esophageal cancer cell lines. Cells were treated with pyrotinib or/and radiation. Cell proliferation, cell cycle distribution, and cell migration were measured. The protein levels involved in cell cycle and DNA repair were measured by Western blot. Results showed that pyrotinib inhibited HER2 activation and exerted an anti-proliferative effect in TE-1 and KYSE30 cells. Furthermore, it enhanced the anti-proliferative effect of radiation in these two cell lines. These effects might be via inhibiting HER2 phosphorylation, inducing G0/G1 arrest, and reducing EMT and DNA repair. Our results indicated that pyrotinib sensitivitied HER2 positive esophageal cancer cells to radiation treatment through various mechanisms. These findings may provide a new therapeutic strategy for treating HER2 positive esophageal cancer.


Subject(s)
Acrylamides/pharmacology , Aminoquinolines/pharmacology , Esophageal Neoplasms/radiotherapy , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Receptor, ErbB-2/antagonists & inhibitors , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/radiation effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Cyclin D1/metabolism , Cyclin-Dependent Kinase 4/metabolism , DNA Repair/drug effects , DNA Repair/radiation effects , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/radiation effects , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/pathology , Humans , Phosphorylation , Receptor, ErbB-2/metabolism , Signal Transduction
6.
J Cancer ; 11(6): 1486-1494, 2020.
Article in English | MEDLINE | ID: mdl-32047555

ABSTRACT

Background: Esophageal squamous cell carcinoma (ESCC) is a common cancer with poor prognosis. The molecular pathogenesis underlying ESCC remains to be explored. Leucine-rich ɑ-2-glycoprotein 1 (LRG1) has been implicated in the pathogenesis of various cancer types, however its role in ESCC is unknown. Materials and Methods: Data from the public database was analyzed to address the expression of LRG1 in ESCC. Gain-of-function studies were performed in select ESCC cell lines by over-expression or addition of recombinant LRG1, while loss-of-function studies achieved by small interfering RNA mediated knockdown. Wound healing and transwell assays were conducted to investigate ESCC cell migration and invasion upon manipulating LRG1 levels. Western blot and Immunofluorescence staining were used to examine the changes in epithelial to mesenchymal transition (EMT) and TGFß signaling pathway. Results: LRG1 mRNA levels were found to be significantly down-regulated in patients with ESCC as well as in several ESCC cell lines. Silencing of LRG1 promoted, while overexpression of LRG1 inhibited ESCC cell migration and invasion. In line with this, Silencing of LRG1 enhanced, while overexpression of LRG1 reduced TGFß signaling and EMT of ESCC cells. Conclusion/Significance: LRG1 suppresses ESCC cell migration and invasion via negative modulation of TGFß signaling and EMT. Down-regulation of LRG1 in ESCC patients may favor tumor metastasis and disease progression.

7.
Invest New Drugs ; 38(2): 500-506, 2020 04.
Article in English | MEDLINE | ID: mdl-31650447

ABSTRACT

Background Esophageal cancer is a very common malignant tumor in China, especially esophageal squamous cell carcinoma (ESCC), but there is currently no effective treatment for patients after first-line chemotherapy failure. Apatinib has shown promising outcomes in treatment with various solid tumors. Objectives To evaluate the clinical efficacy and safety of apatinib combined with S-1 in the treatment of advanced ESCC patients after first-line chemotherapy failure. Methods In this prospective study, fifteen patients with advanced ESCC who failed first-line chemotherapy were enrolled from Nov 2016 to Apr 2019. Patients received the combination therapy with apatinib (250-500 mg, once daily) plus S-1 (40-60 mg based on body surface area, twice daily). Primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), disease control rate (DCR) and objective response rate (ORR). Adverse events (AEs) were recorded to evaluate the safety. Results A total of 12 patients were included in the efficacy analysis. The median PFS was 6.23 months, and the median OS was 8.83 months. Two (16.67%) patients achieved partial remission, 9 patients (75.00%) achieved stable disease and 1 (8.33%) patient achieved progressive disease. DCR and ORR was 91.67%and 16.67%, respectively. Most frequent AEs were hypertension, myelosuppression, weakness, hemorrhage, hand-foot syndrome, total bilirubin elevation, sick, proteinuria, oral ulcer, loss of appetite, and transaminase elevation. The most AEs were in grade I~II. Conclusion The combination therapy of apatinib plus S-1 was effective and well tolerated in the treatment of advanced ESCC patients after first-line chemotherapy failure. The combination therapy has the potential to be a potent therapeutic option for advanced ESCC patients after first-line chemotherapy failure.


Subject(s)
Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Oxonic Acid/therapeutic use , Protein Kinase Inhibitors/therapeutic use , Pyridines/therapeutic use , Tegafur/therapeutic use , Aged , Antineoplastic Agents/adverse effects , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Combinations , Esophageal Neoplasms/mortality , Esophageal Squamous Cell Carcinoma/mortality , Female , Humans , Male , Middle Aged , Oxonic Acid/adverse effects , Protein Kinase Inhibitors/adverse effects , Pyridines/adverse effects , Survival Analysis , Tegafur/adverse effects , Treatment Outcome
8.
Onco Targets Ther ; 12: 2777-2787, 2019.
Article in English | MEDLINE | ID: mdl-31114227

ABSTRACT

Aims: Pyrotinib is a newly developed irreversible pan-ErbB receptor tyrosine kinase inhibitor for treatment of human epidermal growth factor receptor 2 (HER2)-positive cancers, and clinic trials of pyrotinib in treatment of HER2-positive gastric cancer (GC) are underway. Exosomes are tiny vesicles secreted by cancer cells and take essential roles in the progression of carcinoma. Whether pyrotinib application has any effect on the cancer cell-released exosomes has not been studied. The aim of our work was to address if pyrotinib treatment impacts the effect of HER2-positive GC cell-derived exosomes on endothelial cell (EC) progression. Methods: Isolation of exosomes released by HER2-positive NCI-N87 and MKN45 lines after pyrotinib treatment was performed. Then, human umbilical vein endothelial cells (HUVECs) were incubated with different concentrations of exosomes to address their proliferation by 3-(4,5-dimethylthiazol-2-yl)-5-(3-carboxymethoxyphenyl)-2-(4-sulfophenyl)-2H-tetrazolium (MTS). Effect of pyrotinib-treated exosomes at concentration of 10 µg/mL was compared to that without pyrotinib treatment over 96-hr time course. Transwell assay and wound-healing assay were carried out by incubating with exosomes released by NCI-N87 and MKN45 cells with/without pyrotinib treatment over 24-hr time course. The aforementioned experiments were done under same conditions in order to evaluate the combined effect of apatinib and pyrotinib on HUVEC motility and invasive capacity. Results: We showed that HUVEC proliferation, motility and invasive capacity were further enhanced upon incubation with exosomes released by pyrotinib-treated GC cell lines, compared to those without pyrotinib treatment. Significantly, this effect was counteracted by the vascular endothelial growth factor receptor (VEGFR)-2 inhibitor apatinib which inhibits EC progression. Conclusion: Our study suggests that pyrotinib application on HER2-positive GC produces stronger exosomes that promote the proliferation and motility of vascular ECs, and combination of pyrotinib with apatinib provides potentially better therapy.

9.
Sheng Wu Yi Xue Gong Cheng Xue Za Zhi ; 26(2): 334-7, 341, 2009 Apr.
Article in Chinese | MEDLINE | ID: mdl-19499797

ABSTRACT

The co-culture system of early embryos and cancer cells is an important means to observe the biological behavior changes of embryos and cancer cells in vitro. In this study, we co-cultured the 3.5 dpc mouse embryo with malignant tumor cells, investigated the development of blastocyst by observing the hatchment, attachment and outgrowth, observed the biological behavior changes of cancer cells in the embryonic circumstances, and detected the proliferation and apoptosis of cancer cells. Compared with the control, the embryos developed normally in the tumor environments, and the rate of hatchment, attachment and outgrowth increased significantly (P<0.05). However, there was no significant change of cancer cells in morphology, proliferation and apoptosis in the co-culture system (P>0.05). Under the co-culture system, the early embryo developed normally, and the cancer cells also grew well. There may be similarities between the embryos and cancer cell's choice for living. Moreover, the growth of embryos could be promoted by cancer cells in the co-culture system. This might be related to the similarities of gene expression, growth factors and signal transduction mechanisms between embryos and cancer cells.


Subject(s)
Blastocyst/cytology , Embryo Culture Techniques/methods , Embryo, Mammalian , Liver Neoplasms/pathology , Animals , Blastocyst/physiology , Cell Line, Tumor , Coculture Techniques , Embryo, Mammalian/cytology , Humans , Male , Melanoma/pathology , Mice
10.
Hum Gene Ther ; 20(2): 103-11, 2009 Feb.
Article in English | MEDLINE | ID: mdl-18939902

ABSTRACT

Angiogenesis plays an important role in the growth of solid tumors. To date, no information has been acquired on the effectiveness of gene therapy in the orthotopic lung cancer model of syngeneic immunocompetent mice treated with an angiogenesis inhibitor. Here, we report the establishment of such a model in which Lewis lung carcinoma (LL/2) cell suspensions were orthotopically inoculated into the lung parenchyma of C57BL/6 mice, which were also injected with a recombinant adenoviral vector delivering the human endostatin gene (Ad-hE). We found that orthotopic implantation of LL/2 cells into the lung parenchyma produced a solitary tumor nodule in the lung followed by remote mediastinal lymph node metastasis. Conditioned medium from Ad-hE-transfected LL/2 cells apparently inhibited proliferation of human umbilical vein endothelial cells (HUVECs). The level of endostatin protein in serum could be identified by enzyme-linked immunosorbent assay. Treatment with Ad-hE resulted in inhibition of tumor growth and prolongation of survival time of tumor-bearing mice. Immunohistochemical analysis revealed that intratumoral angiogenesis was significantly suppressed. Furthermore, the finding of angiogenesis inhibition was also supported by measuring the number of circulating endothelial cells (CECs). Apoptotic cells were found to be increased within tumor tissues from mice treated with Ad-hE. In addition, treatment with Ad-hE combined with cis-diamminedichloroplatinum(II) (cisplatin) enhanced antitumor activity. These observations provide further evidence of the antitumor effect of endostatin gene therapy, and may be of importance for further exploration of potential application of this combined approach in the treatment of human lung cancer as well as other solid tumors.


Subject(s)
Adenoviridae/genetics , Angiogenesis Inhibitors/therapeutic use , Carcinoma, Lewis Lung/therapy , Endostatins/therapeutic use , Genetic Therapy , Lung Neoplasms/therapy , Animals , Apoptosis , Cell Line, Tumor , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Genetic Vectors/genetics , Humans , Mice , Mice, Inbred C57BL , Recombinant Proteins/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...