Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Nat Commun ; 14(1): 6384, 2023 10 11.
Article in English | MEDLINE | ID: mdl-37821436

ABSTRACT

Currently potential preclinical drugs for the treatment of nonalcoholic steatohepatitis (NASH) and NASH-related pathopoiesis have failed to achieve expected therapeutic efficacy due to the complexity of the pathogenic mechanisms. Here we show Tripartite motif containing 26 (TRIM26) as a critical endogenous suppressor of CCAAT/enhancer binding protein delta (C/EBPδ), and we also confirm that TRIM26 is an C/EBPδ-interacting partner protein that catalyses the ubiquitination degradation of C/EBPδ in hepatocytes. Hepatocyte-specific loss of Trim26 disrupts liver metabolic homeostasis, followed by glucose metabolic disorder, lipid accumulation, increased hepatic inflammation, and fibrosis, and dramatically facilitates NASH-related phenotype progression. Inversely, transgenic Trim26 overexpression attenuates the NASH-associated phenotype in a rodent or rabbit model. We provide mechanistic evidence that, in response to metabolic insults, TRIM26 directly interacts with C/EBPδ and promotes its ubiquitin proteasome degradation. Taken together, our present findings identify TRIM26 as a key suppressor over the course of NASH development.


Subject(s)
Non-alcoholic Fatty Liver Disease , Animals , Rabbits , Non-alcoholic Fatty Liver Disease/genetics , Non-alcoholic Fatty Liver Disease/prevention & control , Signal Transduction , Ubiquitination , Ubiquitin-Protein Ligases/genetics , Ubiquitin-Protein Ligases/metabolism
2.
Adv Sci (Weinh) ; 10(35): e2304521, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37875418

ABSTRACT

The forkhead box transcription factor A2 (FOXA2) is a transcription factor and plays a key role in embryonic development, metabolism homeostasis and tumor cell proliferation; however, its regulatory potential in CRC is not fully understood. Here, it is found that FOXA2 expression is markedly up-regulated in tumor samples of CRC patients as compared with the normal tissues, which is closely associated with the worse survival in patients with CRC. Notably, a positive correlation between FOXA2 and nuclear factor erythroid 2-related factor 2 (Nrf2)/glutathione peroxidase 4 (GPX4) gene expression is observed in CRC patients. Mechanistically, FOXA2 depletion weakens the activation of Nrf2 pathway and decreases GPX4 level in CRC cells, thereby leading to ferroptosis, which is further supported by bioinformatic analysis. More intriguingly, the E3 ubiquitin ligase tripartite motif containing 36 (TRIM36) is identified as a key suppressor of FOXA2, and it is observed that TRIM36 can directly interact with FOXA2 and induce its K48-linked polyubiquitination, resulting in FOXA2 protein degradation in vitro. Taken together, all the studies demonstrate that FOXA2 mediated by TRIM36 promotes CRC progression by inhibiting the Nrf2/GPX4 ferroptosis signaling pathway, thus providing a new therapeutic target for CRC treatment.


Subject(s)
Colorectal Neoplasms , Ferroptosis , Female , Pregnancy , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase , NF-E2-Related Factor 2/genetics , Cell Proliferation/genetics , Colorectal Neoplasms/genetics , Tripartite Motif Proteins , Ubiquitin-Protein Ligases , Hepatocyte Nuclear Factor 3-beta/genetics
3.
Cancer Immunol Immunother ; 72(11): 3727-3738, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37665374

ABSTRACT

Hepatocellular carcinoma (HCC), a prevalent cause of cancer-related deaths, is insensitive to traditional treatments. At different time intervals, the combined antitumor effects of DC-TEX and the programmed death protein 1 (PD-1) antibody (Ab) have not been investigated. In this study, HCC models were established and treated at different time intervals with DC-TEX alone or in combination with PD-1 Ab. In addition, we developed an orthotopic HCC model in BALB/c nude mice and restored T cells. Results demonstrated that the PD-1 + CD8 + T-cell population also increased significantly after DC-TEX treatment, in addition to the increased number of CD8 + T cells. The number of CD8 + T cells increased 72 h after DC-TEX administration. Similar observations were made for PD-1 + CD8 + T cells. Subsequently, PD-1 Ab was administered in combination with DC-TEX at different time points (0, 24, 72, 96, 120, or 168 h). Surprisingly, the combination treatment demonstrated a strong antitumor effect, which was very prominent when PD-1 Ab was administered at 72 h. PD-1 Ab significantly reversed the proliferative ability of PD-1 + CD8 + T cells at 72 h in vitro. The combined antitumor effects of PD-1 Ab and DC-TEX occurred mainly by stimulating CD8 + T cell proliferation and inhibiting T cell exhaustion. In conclusion, our results indicate that the combination of DC-TEX and PD-1 Ab significantly inhibits tumor growth in a murine HCC model and that the timing of PD-1 Ab administration impacts the antitumor effect.


Subject(s)
Carcinoma, Hepatocellular , Exosomes , Liver Neoplasms , Animals , Mice , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/drug therapy , Programmed Cell Death 1 Receptor/metabolism , Exosomes/metabolism , Mice, Nude , CD8-Positive T-Lymphocytes , Dendritic Cells
4.
Iran J Biotechnol ; 21(1): e3405, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36811103

ABSTRACT

Background: Troponin T1 (TNNT1) is implicated in human carcinogenesis. However, the role of TNNT1 in ovarian cancer (OC) remains unclear. Objectives: To investigate the effect of TNNT1 on the progression of ovarian cancer. Materials and Methods: The level of TNNT1 was evaluated in OC patients based on The Cancer Genome Atlas (TCGA). Knockdown or overexpression of TNNT1 using siRNA targeting TNNT1 or plasmid carrying TNNT1 was performed in the ovarian cancer SKOV3 cell, respectively. RT-qPCR was performed to detect mRNA expression. Western blotting was used to examine protein expression. Cell Counting Kit-8, colony formation, cell cycle, and transwell assays were performed to analyze the role of TNNT1 on the proliferation and migration of ovarian cancer. Besides, xenograft model was carried out to evaluate the in vivo effect of TNNT1 on OC progression. Results: Based on available bioinformatics data in TCGA, we found that TNNT1 was overexpressed in ovarian cancer samples comparing to normal samples. Knocking down TNNT1 repressed the migration as well as the proliferation of SKOV3 cells, while overexpression of TNNT1 exhibited opposite effect. In addition, down-regulation of TNNT1 hampered the xenografted tumor growth of SKOV3 cells. Up-regulation of TNNT1 in SKOV3 cells induced the expression of Cyclin E1 and Cyclin D1, promoted cell cycle progression, and also suppressed the activity of Cas-3/Cas-7. Conclusions: In conclusion, TNNT1 overexpression promotes SKOV3 cell growth and tumorigenesis by inhibiting cell apoptosis and accelerating cell-cycle progression. TNNT1 might be a potent biomarker for the treatment of ovarian cancer.

5.
Front Oncol ; 12: 949941, 2022.
Article in English | MEDLINE | ID: mdl-36457494

ABSTRACT

Background: The aim of this study was to retrospectively evaluate the efficacy of full management from first-line to third-line treatments in patients with human epidermal growth factor receptor 2 (Her-2)-negative advanced gastric cancer (GC). Methods: The efficacy and survival time of a total of 126 patients who received the first-line treatment with oxaliplatin plus fluoropyrimidine (S-1 or capecitabine or fluorouracil), the second-line treatment with nab-paclitaxel, and the third-line treatment of immune checkpoint inhibitors between September 2019 and December 2021 were analyzed. Results: A total of 42, 36, and 48 patients received CapeOX, FOLFOX, and SOX as a first-line treatment, respectively. All patients received nab-paclitaxel alone as a second-line treatment. In addition, 31, 56, and 39 patients received nivolumab, sintilimab, and tislelizumab as a third-line treatment, respectively. The median PFS1, median PFS2, and median PFS3 was 6.9 months [95% confidence interval (CI), 6.8-7.4], 5.5 months (95% CI, 5.3-5.7), and 3.5 months (95% CI, 3.4-3.7). The median PFS3 was 3.8 months (95% CI, 3.3-4.2) and 3.5 months (95% CI, 3.3-3.7) among the Epstein-Barr virus (EBV)-positive and EBV-negative, respectively (P = 0.09). In addition, the median PFS3 was 4.2 months (95% CI,3.6-4.7) and 3.5 months (95% CI, 3.3-3.6) in the patients with programmed death ligand 1 (PD-L1) combined positive score (CPS) ≥5 and CPS <5, respectively (P = 0.02). The median OS was 17.4 months (95% CI, 17.2-18.1). The multivariate analysis showed that the two parameters were associated with a significantly longer OS: number of metastatic sites <3 and PD-L1 CPS ≥5. Conclusion: The patients who received three lines of treatment had a long survival time, and the efficacy of immunotherapy was not affected by the EBV subtypes in advanced GC. The toxicity was managed, and the concept of full management needs to be confirmed in the future.

6.
Cell Mol Biol (Noisy-le-grand) ; 67(5): 363-370, 2022 Feb 04.
Article in English | MEDLINE | ID: mdl-35818231

ABSTRACT

The incidence of ocular metastases in patients with disseminated breast cancer is increasing. This study aimed to investigate the clinical features, treatment, and prognosis of breast cancer patients with ocular metastases. For this purpose, a total of 16 patients were diagnosed with ocular metastases. Demographic, treatment, and other clinical data were obtained from patients' charts. The estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor-2 (HER2) statuses of the patients were obtained from the histopathologic reports. Demographic features were analyzed through descriptive statistics, and the Kaplan-Meier method was used for survival analysis. The results showed that among the 16 patients (median age: 41 years), 10 had ER-positive, 8 had PR-positive, and 3 had HER2-positive disease. The choroid was the most commonly involved structure (n = 8). Nine (56%) patients had blurred vision. Treatments for these patients included systemic therapy (six patients), radiotherapy (three patients), and combined therapy (seven patients). The median time from the diagnosis of breast cancer to the diagnosis of ocular metastasis was 52.9 months, and the median time from the diagnosis of metastatic breast cancer at any other site to the diagnosis of ocular metastasis was 21.3 months. The median overall survival (OS) was 136.5 months (95% confidence interval, 40.6-232.4 months), and the median survival duration after ocular metastasis was 32.4 months (95% confidence interval, 20.1-44.7 months). The OS of patients with unilateral eye involvement and bilateral eye involvement did not differ significantly (P = 0.573), nor did the OS of those diagnosed before 2000 and in 2000 or later (P = 0.409). In general, a breast cancer patient with ocular metastasis can have a good prognosis after therapy. However, large-scale clinical studies are needed to confirm our findings.


Subject(s)
Breast Neoplasms , Eye Neoplasms , Adult , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Eye Neoplasms/diagnosis , Eye Neoplasms/secondary , Eye Neoplasms/therapy , Female , Humans , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Retrospective Studies , Survival Analysis
7.
Int J Nanomedicine ; 16: 5513-5529, 2021.
Article in English | MEDLINE | ID: mdl-34429597

ABSTRACT

BACKGROUND: Platinum(IV) complexes with inflammation inhibitory properties are much favored in improving antitumor activities. Nanodrug-delivery system as a preferable measure for antitumor therapy are widely explored in platinum(IV) drug delivery. PURPOSE: The aim for this study was to develop novel bovine serum albumin (BSA) nanoparticles (NPs) based on naproxen platinum(IV) complexes to display a synergistic antitumor mechanism targeting cyclooxygenase-2 (COX-2), metalloproteinase-9 (MMP-9) and inducible nitric oxide synthase (iNOS). METHODS: Herein, we reported the preparation of two BSA NPs of naproxen platinum(IV) complexes, and their antitumor activities were investigated in vitro and in vivo. RESULTS: Both NPs possessed relatively uniform size and good stability for 30 days in aqueous solution. They exhibited prominent antitumor activities in vitro, and showed great potential in reversing drug resistance. Furthermore, these two NPs played superior tumor growth suppression in vivo in contrast to the free compounds, which were comparable to that of cisplatin and oxaliplatin, but induced lower toxic influences than platinum(II) drugs especially to spleen and liver. Moreover, the naproxen platinum(IV) NPs could decrease tumor inflammation targeting COX-2, MMP-9 and iNOs, and decreasing NO production, which would be in favor of enhancing the antitumor competence, and reducing toxicity. CONCLUSION: Taken together, BSA NPs of naproxen platinum(IV) complexes demonstrated a powerful antitumor efficacy in vitro and in vivo. The platinum(IV) NPs with inflammation inhibitory competence targeting multiple enzymes reported in this work afford a new strategy for the development of antitumor therapy to overcome drawbacks of clinical platinum(II) drugs.


Subject(s)
Antineoplastic Agents , Nanoparticles , Animals , Cell Line, Tumor , Humans , Inflammation/drug therapy , Mice , Naproxen , Platinum , Serum Albumin, Bovine
8.
J Cancer ; 9(24): 4627-4634, 2018.
Article in English | MEDLINE | ID: mdl-30588246

ABSTRACT

As a specific subtype of breast cancer, Triple-negative breast cancer (TNBC) is associated with worse prognosis and higher tumor aggressiveness than HER2-amplified or hormone receptor positive breast cancers. Circulating tumor DNA (ctDNA), as a non-invasive "liquid biopsy", is an emerging original blood-based biomarker for early breast cancer diagnosis, monitoring treatment response, and determining prognosis. In TNBC patients, ctDNA has an inherent tendency to characterize tumor heterogeneity and metastasis-specific mutations providing a key alternative to tumor tissue profiling. Several studies have already demonstrated the potential of ctDNA in TNBC patients from early to advanced stages of the disease including diagnosis, therapy decisions and assessment of prognosis. This review provides a critical brief summary of the evidence that gives credence to the utility of ctDNA as a biomarker for its role into clinical management in TNBC.

9.
Oncotarget ; 8(55): 94738-94742, 2017 Nov 07.
Article in English | MEDLINE | ID: mdl-29212262

ABSTRACT

Colorectal cancer (CRC) ranks the fifth leading cause of cancer death in China. EZH2 is a member of Polycomb-group (PcG) family and associated with transcriptional repression and cancer development. In this study, we report the association between a missense variant in EZH2 and risk of CRC. Through a systematic selection of variants in EZH2, we identified rs2302427 in the exon region of EZH2 and genotyped this variant in 852 CRC patients and 1,303 healthy controls using Taqman genotyping assay. The association between this variant and CRC risk was calculated using logistic regression with adjustment of sex, age, smoking status and drinking status. The result showed that rs2302427 was significantly associated with CRC susceptibility under an additive model (P=0.0068). Compared with CC genotype carriers, CG genotype and GG genotype carriers were associated with risk of CRC with odds ratio being 0.78 (95% CI: 0.63-0.96, P=0.0198) and 0.54 (95% CI: 0.24-1.18, P=0.1224), respectively. When stratified by sex, age, smoking status or drinking status, significant associations were observed only in younger individuals (OR=0.67, 95% CI: 0.50-0.89, P=0.0067) or smokers (OR=0.65, 95% CI: 0.48-0.88, P=0.0051). This study provides new insights into the personalized prevention of colorectal cancer.

10.
Onco Targets Ther ; 9: 3901-6, 2016.
Article in English | MEDLINE | ID: mdl-27418834

ABSTRACT

This study was conducted to evaluate the toxicity and efficacy of pemetrexed plus dendritic cells (DCs) when administered as third-line treatment for metastatic esophageal squamous cell carcinoma (ESCC). All patients in the study group had previously failed first-line treatment with 5-fluorouracil and cisplatin-based regimens, as well as second-line treatment with taxane-based regimens. A total of 31 patients were treated with pemetrexed (500 mg/m(2)) plus DCs on day 1, every 3 weeks. DCs were given for one cycle of 21 days. Thirty patients were evaluated for their response. No patient had a complete response, three patients (10.0%) had a partial response, ten patients (33.3%) had stable disease, and 17 patients (56.7%) had progressive disease. The overall response rate was 10.0%. The median progression-free survival (PFS) time was 2.9 months (95% CI, 2.7-3.2), and the median overall survival (OS) time was 7.1 months (95% CI, 6.4-7.9). The median PFS and OS times among patients with high and low levels of miR-143 expression in their blood serum were significantly different: median PFS times =3.2 months (95% CI, 2.9-3.4) and 2.7 months (95% CI, 2.4-3.0), respectively (P=0.017), and median OS times =7.8 months (95% CI, 6.8-8.9) and 6.3 months (95% CI, 5.3-7.3), respectively (P=0.036). No patient experienced Grade 4 toxicity. Combined third-line treatment with pemetrexed and DCs was marginally effective and well tolerated in patients with advanced ESCC. Serum miR-143 levels are a potential biomarker for predicting the efficacy of pemetrexed plus DCs in the treatment of ESCC.

11.
Transl Res ; 170: 1-7, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26687391

ABSTRACT

This study was conducted to evaluate microRNAs (miRNAs) as biomarkers for use in predicting the efficacy of maintenance therapy with pemetrexed in patients with stage IIIb or IV lung adenocarcinoma and who had already received first-line treatment with pemetrexed plus platinum. Patients who were negative for epidermal growth factor receptor (EGFR) mutations or anaplastic lymphoma kinase (ALK) translocations were assigned to a pemetrexed group and an observation group. Patients in the pemetrexed group (n = 76) received maintenance treatment with pemetrexed (500 mg/m(2), once every 21 days) plus best supportive care. Patients in the observation group (n = 72) agreed to receive only best supportive care until disease progression. Blood samples were collected from all patients in both groups before treatment and were used to detect expression levels of various miRNAs in serum by the Reverse Transcription-Polymerase Chain Reaction (RT-PCR) method. The expression levels of miR-25, miR-145, and miR-210 were significantly different in the 2 groups of patients. Furthermore, the median progression-free survival (PFS) times for patients in the pemetrexed and observation groups were 4.5 and 2.9 months, respectively. The PFS times among patients in the pemetrexed group varied significantly and were related to patient expression levels of miR-25, miR-145, and miR-210, whereas patients in the observation group showed no differences in PFS time. Our data suggest miR-25, miR-145, and miR-210 as predictors for the efficacy of maintenance treatment with pemetrexed in lung adenocarcinoma patients who were negative for EGFR mutations or ALK translocations.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/genetics , Lung Neoplasms/drug therapy , Lung Neoplasms/genetics , MicroRNAs/genetics , Pemetrexed/therapeutic use , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aged , Anaplastic Lymphoma Kinase , Biomarkers, Tumor/genetics , Disease-Free Survival , ErbB Receptors/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Mutation , Receptor Protein-Tyrosine Kinases/genetics , Treatment Outcome
12.
Sci Rep ; 5: 10244, 2015 Jul 13.
Article in English | MEDLINE | ID: mdl-26165253

ABSTRACT

Tumour necrosis factor-α (TNF-α) is critical in the regulation of inflammation and tumour progression. TNF-α-308G > A is associated with constitutively elevated TNF-α expression. The purpose of this study was to assess the association between TNF-α-308G > A and breast cancer (BC) risk by subtype and the connection between genotypes and clinical features of BC. A total of 768 patients and 565 controls were enrolled in this study, and genotypes were detected using the TaqMan assay. No effect on susceptibility for any BC subtype was found for the TNF-α-308 polymorphism in our study or in the pooled meta-analysis. This polymorphism was shown to be associated with age at menarche in all BC and in progesterone receptor-negative BC. Interestingly, triple negative breast cancer (TNBC) patients with TNF-α-308A had an increased risk of distant tumour metastasis (OR = 3.80, 95% CI: 1.31-11.02, P = 0.009). Multi-regression analysis showed that TNF-α-308A was also a risk factor for distant tumour metastasis after adjustment for tumour size and lymph node metastasis status (OR = 6.26, 95% CI: 1.88-20.87, P = 0.003). These findings indicate that TNF-α might play a distinct role in the progression of TNBC, especially in distant tumour metastasis of TNBC.


Subject(s)
Triple Negative Breast Neoplasms/genetics , Tumor Necrosis Factor-alpha/genetics , Adult , Alleles , Asian People/genetics , Databases, Factual , Female , Genotype , Humans , Middle Aged , Neoplasm Metastasis , Odds Ratio , Polymorphism, Single Nucleotide , Regression Analysis , White People/genetics
13.
Med Oncol ; 31(8): 63, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24958515

ABSTRACT

The aim of this study was to determine the efficacy and toxicity of pemetrexed plus dendritic cells (DCs) in patients suffering from stage IIIB or IV lung adenocarcinoma, who had undergone maintenance treatment with gefitinib or erlotinib. Patients who had failed gefitinib or erlotinib maintenance treatment had ECOG performance statuses ranging from 0 to 2.27 patients received pemetrexed plus DCs as second-line treatment. Dosage: 500 mg/m(2) pemetrexed was administered on day 1 of a 21-day cycle. DCs were given for one cycle of 21 days. Three patients (11.1 %) experienced a partial response and 14 patients (51.9 %) showed stable disease. Ten patients (37.0 %) had progressive disease. The median time to progression-free survival (PFS) was 4.8 months [95 % confidence interval (CI) 4.4-5.2], and the median overall survival was 10.7 months (95 % CI 10.3-11.2). In the subgroup analysis, PFS had a significant difference between the low ratio of CD4/CD8 and normal ratio of CD4/CD8, with 4.5 months (95 % CI 4.2-4.9) and 5.0 months (95 % CI 4.5-5.7), (Log Rank = 0.039), respectively. No one patient experienced grade 4 toxicity. A regimen of pemetrexed combined with DCs is marginally effective and well tolerated in patients with stage IIIB or IV lung adenocarcinoma who had received gefitinib or erlotinib first-line treatment.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Dendritic Cells/transplantation , Enzyme Inhibitors/therapeutic use , Glutamates/therapeutic use , Guanine/analogs & derivatives , Lung Neoplasms/drug therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Adenocarcinoma/therapy , Adenocarcinoma of Lung , Adult , Aged , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/therapy , Disease-Free Survival , Enzyme Inhibitors/adverse effects , Female , Glutamates/adverse effects , Guanine/adverse effects , Guanine/therapeutic use , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Kaplan-Meier Estimate , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Lung Neoplasms/therapy , Male , Middle Aged , Pemetrexed , Treatment Outcome
14.
J Immunother ; 37(4): 250-5, 2014 May.
Article in English | MEDLINE | ID: mdl-24714359

ABSTRACT

The aim of this study was to evaluate the safety and effectiveness of erlotinib plus DC/CIK in maintenance therapy of advanced non-small cell lung cancer. After 4 cycles of the 2-drug regimen treatment with platinum, the 54 patients with non-small cell lung cancer in phase IIIb or IV reached stable or beyond stable stages. The patients were then randomly divided into 2 groups. One group was treated with erlotinib therapy (erlotinib group), and the other was treated with DC/CIK plus erlotinib (DC/CIK plus erlotinib group). The progression-free survival of the erlotinib group and the DC/CIK plus erlotinib group was 3.98 months (95% CI, 3.56-4.40) and 5.02 months (95% CI, 4.32-5.72) (P=0.002), respectively. The median overall survival of the erlotinib group and the DC/CIK plus erlotinib group was 9.9 months (95% CI, 9.1-10.6) and 10.5 months (95% CI, 9.6-11.4) (P=0.29), respectively. The levels of CD3, CD4, and CD8 were significantly different before and after the treatment in the DC/CIK plus erlotinib group, but not in the erlotinib group. There was no significant difference in toxicity between the 2 groups. In conclusion, there was no statistically significant difference in overall survival between DC/CIK plus erlotinib and erlotinib as maintenance therapy. DC/CIK plus erlotinib was well tolerated with a manageable safety profile.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Cytokine-Induced Killer Cells/transplantation , Dendritic Cells/transplantation , Immunotherapy, Adoptive , Lung Neoplasms/therapy , Protein Kinase Inhibitors/therapeutic use , Quinazolines/therapeutic use , Aged , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Combined Modality Therapy , Erlotinib Hydrochloride , Female , Humans , Immunotherapy, Adoptive/adverse effects , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Metastasis , Neoplasm Staging , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Quinazolines/administration & dosage , Quinazolines/adverse effects , Risk Factors , Treatment Outcome
15.
Chin Med J (Engl) ; 126(22): 4277-81, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24238512

ABSTRACT

BACKGROUND: Famitinib is a novel and potent multitargeting receptor tyrosine kinase inhibitor. The phase I clinical study showed that famitinib was well tolerated and had a broad anti-tumor spectrum. The purpose of this study was to examine the efficacy and safety of famitinib for the treatment of metastatic renal cell carcinoma (mRCC). METHODS: The data of famitinib in treating patients with mRCC from the single-center phases I and II clinical trials were analyzed. Famitinib was administered orally at the dose of 13-30 mg once daily until tumor progression, occurrence of intolerable adverse reactions or withdrawal of the informed consent. RESULTS: A total of 24 patients with mRCC were treated including 17 patients at a dose of 25 mg once daily, 4 patients at a dose of 27 mg and 1 patient each at a dose of 13 mg, 20 mg and 30 mg, respectively. Twelve (50.0%) patients achieved partial response (PR) and 9 patients achieved stable disease (SD). Progressive disease was found in 3 (12.5%) patients. The disease control rate was 87.5%. The median follow-up time was 17.6 months; the median progression free survival (PFS) was 10.7 (95% CI 7.0-14.4) months; and the estimated median overall survival (OS) time was 33.0 (95% CI 8.7-57.3) months. The adverse drug reactions mainly included hypertension (54.1%), hand-foot skin reactions (45.8%), diarrhea (33.3%), mucositis (29.2%), neutropenia (45.8%), thrombocytopenia (29.2%), hyperlipidemia (41.7%) and proteinuria (41.7%). The incidence rate of grades 3 and 4 adverse events was low, mainly including hypertension 12.5%, hand-foot skin reactions 4.2%, neutropenia 4.2%, thrombocytopenia 4.2%, hyperlipidemia 4.2% and proteinuria 12.5%. CONCLUSIONS: Famitinib has significant anti-tumor activity in mRCC. The common adverse reactions are generally manageable.


Subject(s)
Carcinoma, Renal Cell/drug therapy , Indoles/therapeutic use , Kidney Neoplasms/drug therapy , Pyrroles/therapeutic use , Adult , Aged , Female , Humans , Indoles/adverse effects , Male , Protein Kinase Inhibitors , Pyrroles/adverse effects , Retrospective Studies , Treatment Outcome , Young Adult
16.
Cancer Chemother Pharmacol ; 72(5): 1043-53, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24043137

ABSTRACT

PURPOSE: To evaluate the safety, tolerability, pharmacokinetics and antitumor activities of famitinib (famitinib L-malate), a novel oral multitargeting tyrosine kinase inhibitor that acts against vascular endothelial growth factor receptor-2, platelet-derived growth factor receptor, stem cell factor receptor (c-kit), FMS-like tyrosine kinase-3 receptor and protooncogene tyrosine kinase receptor in patients with advanced solid cancer. METHODS: Patients received once daily oral famitinib. Doses were increased from 4 to 8, 13, 20, 27, 24, 25 and eventually 30 mg. Each cycle was defined as 28 days. The pharmacokinetic profile and various biomarkers were evaluated during the first cycle. Antitumor efficacy was evaluated every 8 weeks. RESULTS: Fifty-four patients were evaluable for safety and efficacy. Dose-limiting toxicities were observed in 2 of 3 patients at 30 mg. The dose-limiting toxicities observed in the first cycle of famitinib treatment included hypertension, hand-foot skin reaction and diarrhea. Grade 3 hypertriglyceridemia/hypercholesterolemia and proteinuria were notable side effects in the subsequent treatment cycles. Other common side effects included bone marrow suppression, oral mucositis, fatigue, pain, elevated transaminase or bilirubin, peripheral sensory disturbance and hypothyroidism, most of which were mild to moderate in severity. Pharmacokinetic studies revealed no significant accumulation of famitinib or its major metabolite, M3. The half-lives of famitinib and M3 were approximately 28.7-33.8 and 41.3-47.7 h, respectively. Food demonstrated a minimal effect on the pharmacokinetics of famitinib. Eight partial responses were determined, including 6 cases of renal cell carcinoma, 1 case of gastrointestinal stromal tumor (GIST) and 1 case of alveolar soft part sarcoma. Fourteen patients demonstrated stable disease with various degrees of tumor shrinkage. CONCLUSIONS: Famitinib is generally well tolerated. Famitinib demonstrates a wide spectrum of antitumor activities, which warrants further study in renal cell carcinoma, GIST, hepatocellular carcinoma and soft tissue sarcoma. The recommended dose for future phase II clinical trials is 25 mg.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Indoles/therapeutic use , Neoplasms/drug therapy , Protein Kinase Inhibitors/therapeutic use , Pyrroles/therapeutic use , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Adult , Aged , Angiogenesis Inhibitors/administration & dosage , Angiogenesis Inhibitors/adverse effects , Angiogenesis Inhibitors/pharmacokinetics , Biotransformation , Carcinoma, Renal Cell/blood , Carcinoma, Renal Cell/drug therapy , China/epidemiology , Cohort Studies , Dose-Response Relationship, Drug , Female , Half-Life , Humans , Hypertension/chemically induced , Hypertension/epidemiology , Incidence , Indoles/administration & dosage , Indoles/adverse effects , Indoles/pharmacokinetics , Kidney Neoplasms/blood , Kidney Neoplasms/drug therapy , Male , Middle Aged , Neoplasms/blood , Neoplasms/metabolism , Postprandial Period , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/pharmacokinetics , Pyrroles/administration & dosage , Pyrroles/adverse effects , Pyrroles/pharmacokinetics , Sarcoma/blood , Sarcoma/drug therapy , Young Adult
17.
Med Oncol ; 30(2): 550, 2013.
Article in English | MEDLINE | ID: mdl-23576138

ABSTRACT

To determine the efficacy and toxicity of Pemetrexed plus Oxaliplatin in patients suffering from stage IIIb or IV lung adenocarcinoma and being treated with Erlotinib as second-line treatment, a total of 45 patients were randomly divided into two groups. One group was treated with 500 mg/m(2) Pemetrexed plus 100 mg/m(2) Oxaliplatin, and the other was treated with 500 mg/m(2) Pemetrexed plus 75 mg/m(2) Cisplatin. All drugs were administered on day one of a 21-day cycle. In the Oxaliplatin group, 3 patients (13.6 %) experienced partial response (PR), 9 patients (41.0 %) showed stable disease (SD), and 10 patients (45.5 %) had progressive disease (PD). In the Cisplatin group, 2 patients (8.7 %) experienced PR, 7 patients (30.4 %) showed SD, and 14 patients (60.9 %) had PD. The PFS of the Oxaliplatin group and the Cisplatin group was 4.45 months (95 % CI 4.10-4.80) and 3.96 months (95 % CI 3.68-4.24) (P = 0.03), respectively. The median overall survival (OS) was 10.8 months (95 % CI 10.2-11.5) and 10.7 months (95 % CI 10.2-11.3) (P = 0.72), respectively. There was no statistically significant difference in the occurrence rate of grades 3 and 4 myelotoxicity between the two groups. However, there was a significant difference in the occurrence rate of grades 3 and 4 gastrointestinal reactions and peripheral neurotoxicity between the two groups (P < 0.05). A regime combining Pemetrexed and Oxaliplatin was marginally effective and well tolerated in patients with stage IIIb or IV lung adenocarcinoma who have received Erlotinib as second-line treatment.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Lung Neoplasms/drug therapy , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cisplatin/administration & dosage , Cisplatin/adverse effects , Disease-Free Survival , Erlotinib Hydrochloride , Female , Glutamates/administration & dosage , Glutamates/adverse effects , Guanine/administration & dosage , Guanine/adverse effects , Guanine/analogs & derivatives , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Organoplatinum Compounds/administration & dosage , Organoplatinum Compounds/adverse effects , Oxaliplatin , Pemetrexed , Quinazolines/adverse effects , Quinazolines/therapeutic use , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...