Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 48
Filter
1.
Int J Surg ; 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38652147

ABSTRACT

BACKGROUND: We aimed to compare combined intraoperative chemotherapy and surgical resection with curative surgical resection alone in colorectal cancer patients. METHODS: We performed a multicenter, open-label, randomized, phase III trial. All eligible patients were randomized and assigned to intraoperative chemotherapy and curative surgical resection or curative surgical resection alone (1:1). Survival actualization after long-term follow-up was performed in patients analyzed on an intention-to-treat basis. RESULTS: From January 2011 to January 2016, 696 colorectal cancer patients were enrolled and randomly assigned to intraoperative chemotherapy and radical surgical resection (n=341) or curative surgical resection alone (n=344). Intraoperative chemotherapy with surgical resection showed no significant survival benefit over surgical resection alone in colorectal cancer patients (3-year DFS: 91.1% vs. 90.0%, P=0.328; 3-year OS: 94.4% vs. 95.9%, P=0.756). However, colon cancer patients benefitted from intraoperative chemotherapy, with a relative 4% reduction in liver and peritoneal metastasis (HR=0.336, 95% CI: 0.148-0.759, P=0.015) and a 6.5% improvement in 3-year DFS (HR=0.579, 95% CI: 0.353-0.949, P=0.032). Meanwhile, patients with colon cancer and abnormal pretreatment CEA levels achieved significant survival benefits from intraoperative chemotherapy (DFS: HR=0.464, 95% CI: 0.233-0.921, P=0.029 and OS: (HR=0.476, 95% CI: 0.223-1.017, P=0.049). CONCLUSIONS: Intraoperative chemotherapy showed no significant extra prognostic benefit in total colorectal cancer patients who underwent radical surgical resection; however, in colon cancer patients with abnormal pretreatment serum CEA levels (> 5 ng/ml), intraoperative chemotherapy could improve long-term survival.

2.
J Pharm Biomed Anal ; 229: 115355, 2023 May 30.
Article in English | MEDLINE | ID: mdl-37018958

ABSTRACT

OBJECTIVE: To investigate the metabolic mechanisms of Chinese and Western medicines on the metabolic network of striatal injury in a copper-loaded rat model of Wilson disease (WD) from a metabolomic perspective. METHODS: We divided 60 rats into 4 groups of 15 rats each according to a random number table, namely the control group, the model group, the Bushen Huoxue Huazhuo Recipe group, and the penicillamine group, and subsequently replicated the WD copper-loaded rat model according to the literature method for a total of 12 weeks. From the 7th week onwards, each intervention group was given an equivalent dose of the corresponding drug, and the control and model groups were given an equal volume of saline gavage until the end of the model replication. We used 1H NMR metabolomics techniques combined with multivariate statistical methods to describe the changes in the striatal metabolic profile of nerve injury in Wilson's disease and to analyze the effect of different treatments on their biomarker interventions. RESULTS: Nerve cell damage was evident in the WD copper-loaded rat model and could be reduced to varying degrees by different methods of intervention in the striatal nerve cells. The content of glycine, serine metabolism, and valine metabolism decreased in WD copper-loaded rat model; aspartate content increased after penicillamine intervention; glycolytic metabolism, valine metabolism, taurine metabolism, and tyrosine metabolism increased in the group of Bushen Huoxue Huazhuo Recipe. CONCLUSION: Different intervention methods of Chinese and Western medicine affect aspartate, glycolysis, taurine, tyrosine, valine, and carbon metabolism in striatal tissues of WD copper-loaded rats, and can regulate the metabolism of small molecules, which in turn have certain repairing effects on nerve damage in WD copper-loaded rats.


Subject(s)
Hepatolenticular Degeneration , Rats , Animals , Hepatolenticular Degeneration/drug therapy , Hepatolenticular Degeneration/metabolism , Copper , Proton Magnetic Resonance Spectroscopy , Aspartic Acid , Penicillamine/pharmacology , Penicillamine/therapeutic use , Metabolomics
3.
Lancet Gastroenterol Hepatol ; 8(5): 422-431, 2023 05.
Article in English | MEDLINE | ID: mdl-36870360

ABSTRACT

BACKGROUND: The current standard treatment for locally advanced rectal cancer is neoadjuvant chemoradiotherapy followed by radical surgery, but this approach can lead to multiple complications. We aimed to investigate the clinical activity and safety of neoadjuvant therapy with sintilimab, a single-agent PD-1 antibody, in patients with mismatch-repair deficient locally advanced rectal cancer. METHODS: This open-label, single-arm, phase 2 study was done at the Sun Yat-sen University Cancer Center, Guangzhou, China. Patients aged 18-75 years with mismatch-repair deficient or microsatellite instability-high locally advanced rectal cancer were enrolled and received neoadjuvant sintilimab monotherapy (200 mg by intravenous infusion) every 21 days. After an initial four cycles of treatment, patients and clinicians could choose one of the following options: total mesorectal excision surgery, followed by four cycles of adjuvant sintilimab with or without CapeOX chemotherapy (capecitabine 1000 mg/m2, orally administered twice daily on days 1-14; oxaliplatin 130 mg/m2, intravenously administered on day 1 every 3 weeks), determined by clinicians; or another four cycles of sintilimab followed by radical surgery or observation (only for patients with a clinical complete response; also known as the watch and wait strategy). The primary endpoint was the complete response rate, which included both a pathological complete response after surgery and a clinical complete response after completion of sintilimab treatment. Clinical response was evaluated by digital rectal examination, MRI, and endoscopy. Response was assessed in all patients who received treatment at least until the first tumour response assessment, after the first two cycles of sintilimab. Safety was analysed in all patients who received at least one dose of treatment. This trial is closed to enrolment and is registered with ClinicalTrials.gov (NCT04304209). FINDINGS: Between Oct 19, 2019, and June 18, 2022, 17 patients were enrolled and received at least one dose of sintilimab. The median age was 50 years (IQR 35-59) and 11 (65%) of 17 patients were male. One patient was excluded from efficacy analyses because they were lost to follow-up after the first sintilimab cycle. Of the remaining 16 patients, six underwent surgery, of whom three had a pathological complete response. Nine other patients had a clinical complete response and chose the watch and wait strategy. One patient had a serious adverse event and discontinued treatment; this patient did not have a complete clinical response and refused to undergo surgery. A complete response was thus noted for 12 (75%; 95% CI 47-92) of 16 patients. One of the three patients who underwent surgery but did not have a pathological complete response showed an increase in tumour volume after the initial four cycles of sintilimab (at which point they underwent surgery); this patient was deemed to have primary resistance to immune checkpoint inhibitors. After a median follow-up of 17·2 (IQR 8·2-28·5) months, all patients were alive and none had disease recurrence. Only one (6%) patient had a grade 3-4 adverse event, which was deemed a serious adverse event (grade 3 encephalitis). INTERPRETATION: The preliminary results of this study suggest that anti-PD-1 monotherapy is effective and tolerable for patients with mismatch-repair deficient locally advanced rectal cancer and could potentially spare some patients from radical surgery. Longer treatment courses might be needed to achieve maximum effects in some patients. Longer follow-up is also needed to observe the duration of response. FUNDING: The National Natural Science Foundation of China, CAMS Innovation Fund for Medical Sciences, Science and Technology Program of Guangzhou, and Innovent Biologics.


Subject(s)
Neoadjuvant Therapy , Rectal Neoplasms , Female , Humans , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Neoplasm Recurrence, Local/pathology , Neoplasm Staging , Rectal Neoplasms/drug therapy , Treatment Outcome
4.
Front Immunol ; 13: 1010490, 2022.
Article in English | MEDLINE | ID: mdl-36325347

ABSTRACT

Currently, immune checkpoint inhibitors (ICIs) are the mainstay of treatment for Lynch syndrome patients. However, the tumor regression features in radiology and pathology are inconsistent for patients who are treated with ICIs, which sometimes confuses surgical decision-making. Here, we report a case in which a 36-year-old patient suffering from infertility was diagnosed with Lynch syndrome-associated synchronous endometrial cancer and colon cancer, and persistently enlarged left iliac paravascular lymph nodes were detected after receiving sintilimab treatment, a programmed cell death 1 (PD-1) receptor inhibitor. Fortunately, when she was about to undergo hysterectomy and bilateral salpingo-oophorectomy, intraoperative pathology examination did not reveal any cancer cells in these lymph nodes, and therefore, her reproductive organs were preserved. Later, the patient successfully conceived and gave birth to a healthy male neonate with no immune-related adverse events (irAEs) during an 11-month follow-up. This case indicates that surgeons should carefully inspect the imaging characteristics after immunotherapy and that organ preservation is possible even for patients who fail to achieve complete clinical regression, which is especially important for female patients of childbearing age.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms, Hereditary Nonpolyposis , Endometrial Neoplasms , Infertility , Humans , Infant, Newborn , Male , Female , Pregnancy , Adult , Colorectal Neoplasms, Hereditary Nonpolyposis/complications , Programmed Cell Death 1 Receptor , Organ Preservation , Endometrial Neoplasms/complications , Endometrial Neoplasms/pathology , Colonic Neoplasms/complications , Genitalia
5.
Cell Death Dis ; 13(5): 439, 2022 05 06.
Article in English | MEDLINE | ID: mdl-35523764

ABSTRACT

According to the EPOC study, chemotherapy could improve 5-year disease-free survival of stage IV colon cancer patients by 8.1%. However, more molecular biomarkers are required to identify patients who need neoadjuvant chemotherapy. DENND2D expression was evaluated by immunohistochemistry in 181 stage IV colon cancer patients. The prognosis was better for patients with DENND2D expression than patients without DENND2D expression (5-year overall survival [OS]: 42% vs. 12%, p = 0.038; 5-year disease-free survival: 20% vs. 10%, p = 0.001). Subgroup analysis of the DENND2D-negative group showed that patients treated with neoadjuvant chemotherapy achieved longer OS than patients without neoadjuvant chemotherapy (RR = 0.179; 95% CI = 0.054-0.598; p = 0.003). DENND2D suppressed CRC proliferation in vitro and in vivo. Downregulation of DENND2D also promoted metastasis to distant organs in vivo. Mechanistically, DENND2D suppressed the MAPK pathway in CRC. Colon cancer patients who were DENND2D negative always showed a worse prognosis and were more likely to benefit from neoadjuvant chemotherapy. DENND2D may be a new prognostic factor and a predictor of the need for neoadjuvant chemotherapy in stage IV colon cancer.


Subject(s)
Colonic Neoplasms , Neoadjuvant Therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Disease-Free Survival , Humans , Immunohistochemistry , Neoplasm Staging , Retrospective Studies
6.
Bioresour Technol ; 355: 127265, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35526714

ABSTRACT

p-Nitrophenol is usually present in ammonia-rich wastewaters produced by some chemical plants. In this work, the response of anammox process to long-term p-nitrophenol stress was investigated. The changes in the efficiency, sludge characteristics, and microorganisms of the anammox system under different levels of p-nitrophenol stress were examined, and the potential stress mechanisms of p-nitrophenol on anammox were further speculated. The results showed that 10-50 mg/L p-nitrophenol had no obvious impact on nitrogen removal efficiency, but stimulated the secretion of more extracellular polymeric substances. 60 mg/L p-nitrophenol caused the nitrogen removal efficiency to decrease by 64.5% in 5 days. Long-term exposure to p-nitrophenol led to 8.6% reduction in Candidatus_Kuenenia abundance and 18.4%-35.9% decrease in the expression level of anammox bacterial functional genes. Molecular simulation indicated that p-nitrophenol could bind to key enzymes of anammox. This study provides new insights into the treatment of wastewater containing p-nitrophenol or phenol by anammox.


Subject(s)
Anaerobic Ammonia Oxidation , Bioreactors , Anaerobiosis , Bioreactors/microbiology , Denitrification , Nitrogen/analysis , Nitrophenols , Oxidation-Reduction , Sewage , Wastewater
7.
J Immunother Cancer ; 9(2)2021 02.
Article in English | MEDLINE | ID: mdl-33579737

ABSTRACT

BACKGROUND: CKLF-like MARVEL transmembrane domain-containing 6 (CMTM6), a programmed death-ligand 1 (PD-L1) regulator, is widely expressed in various tumors and regulates the immune microenvironment. However, its prognostic value remains controversial, and the roles of CMTM6 in colorectal cancer (CRC) are still unknown. In this study, we aimed to elaborate the expression patterns of CMTM6 and PD-L1 in CRC and investigate their relationship with the infiltration of T cells and the prognosis of patients with CRC. METHODS: Analysis of CMTM6 mRNA levels, gene ontology enrichment analysis and single-sample gene set enrichment analysis were performed in a The Cancer Genome Atlas colon cancer cohort. The expression of CMTM6 and PD-L1 and the infiltration of T cells in tumor tissues from our cohort containing 156 patients with CRC receiving adjuvant chemotherapy and 77 patients with CRC without chemotherapy were examined by immunohistochemistry assay. RESULTS: CMTM6 expression was upregulated in CRC compared with normal colon tissues, and CMTM6 levels were lower in advanced tumors than in early-stage tumors. High expression of CMTM6 correlated with lower pT stage and more CD4+/CD8+ tumor-infiltrating lymphocytes (TILs) and predicted a favorable prognosis in CRC. PD-L1 was expressed in CRC tissues at a low level, and PD-L1 positivity in tumor stroma (PD-L1(TS)), but not PD-L1 positivity in cancer cells (PD-L1(CC)), was associated with an increased density of CD4+ TILs and a favorable prognosis. The coexpression status of CMTM6 and PD-L1(TS) divided patients with CRC into three groups with low, moderate and high risks of progression and death, and patients with CMTM6High/PD-L1(TS)+ status had the longest survival. Moreover, the prognostic value of CMTM6/PD-L1 expression was more significant in patients with CRC treated with adjuvant chemotherapy than in those not treated with chemotherapy. CONCLUSION: CMTM6 has a critical impact on the immune microenvironment and can be used as an independent prognostic factor for CRC. The coexpression status of CMTM6 and PD-L1 can be used as a new classification to stratify the risk of progression and death for patients with CRC, especially for patients receiving adjuvant chemotherapy. These findings may provide insights into improving responses to immunotherapy-included comprehensive treatment for CRC in the future.


Subject(s)
B7-H1 Antigen/genetics , Colorectal Neoplasms/genetics , Gene Expression Profiling/methods , Lymphocytes, Tumor-Infiltrating/immunology , MARVEL Domain-Containing Proteins/genetics , Myelin Proteins/genetics , Adult , Aged , Aged, 80 and over , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/immunology , Colorectal Neoplasms/pathology , Female , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/pathology , Male , Middle Aged , Neoplasm Staging , Prognosis , Sequence Analysis, RNA , Survival Analysis , Up-Regulation
8.
Cancer Commun (Lond) ; 40(8): 355-369, 2020 08.
Article in English | MEDLINE | ID: mdl-32609436

ABSTRACT

BACKGROUND: Hepatocellular carcinoma (HCC) is a major health problem and a primary cause of cancer-related death worldwide. Although great advances have achieved recently by large-scale high-throughput analysis, the precise molecular mechanism underlying HCC progression remains to be clearly elucidated. We investigated the relationship between Tescalcin (TESC), a candidate oncogene, and clinicopathological features of HCC patients and explored the role of TECS in HCC development. METHODS: To identify new genes involved in HCC development, we analyzed The Cancer Genome Atlas liver cancer database, and TESC was selected for further investigation. HCC tissue microarray analysis for TESC and its association with clinicopathological features were performed to investigate its clinical significance. TESC was knocked down by using short-hairpin RNAs. Cell proliferation was analyzed by WST-1 assay and cell counting. Cell apoptosis was tested by fluorescence-activated cell sorting. A subcutaneous xenograft tumor model in nude mice was established to determine the in vivo function of TESC. Affymetrix microarray was used to identify its molecular mechanism. RESULTS: TESC was significantly increased in HCC tissues compared with the adjacent normal liver tissues. High expression of TESC was detected in 61 of 172 HCC patients by tissue microarray. Large tumor (> 5 cm) and elevated total bilirubin were associated with high TESC expression (both P < 0.050). In multivariate analysis, TESC was identified as an independent prognostic factor for short overall survival of HCC patients. TESC knockdown impaired HCC cell growth in vitro and in vivo. TESC knockdown significantly increased cell apoptosis in HCC cell lines. Furthermore, Affymetrix microarray analysis revealed that TESC knockdown inhibited tumor proliferation-related pathways while activated cell death-related pathways. CONCLUSION: TESC was identified as an independent prognostic factor for short overall survival of HCC patients, and was critical for HCC cell proliferation and survival.


Subject(s)
Calcium-Binding Proteins/genetics , Carcinoma, Hepatocellular , Chemoembolization, Therapeutic , Liver Neoplasms , Animals , Apoptosis , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/pathology , Cell Proliferation , Female , Gene Knockdown Techniques , Humans , Liver Neoplasms/diagnosis , Liver Neoplasms/pathology , Male , Mice , Mice, Nude , Neoplasm Transplantation , Prognosis
9.
Am J Cancer Res ; 10(5): 1467-1476, 2020.
Article in English | MEDLINE | ID: mdl-32509392

ABSTRACT

Skin toxicity, especially hand-foot syndrome (HFS), is one of the most common sorafenib-induced adverse events (AEs) in hepatocellular carcinoma (HCC) patients, leading to treatment interruption and failure. Mucocutaneous inflammation may cause HFS; therefore, we investigated whether celecoxib can alleviate HFS, improve patients' quality of life and increase survival when administered in conjunction with active therapy. Our randomized, open-label study prospectively enrolled 116 advanced HCC patients receiving sorafenib as targeted therapy from July 2015 to July 2016. All patients were randomly assigned (1:1) via a computer-generated sequence to receive sorafenib with or without celecoxib. Sorafenib-related AEs were recorded, Survival was compared between the two groups. Compared to the Sorafenib group, the SoraCele group had lower incidence rates of ≥ grade 2 and grade 3 HFS (63.8% vs 29.3%, P < 0.001; 19.0% vs 3.4%, P = 0.008, respectively), hair loss, rash and abdominal pain. Kaplan-Meier analysis revealed a lower risk of ≥ grade 2 HFS (HR, 0.384; P = 0.002) and a lower dose reduction/interruption rate (46.6% to 15.5%, P < 0.001) in the SoraCele group. Cox proportional hazards regression analysis demonstrated that celecoxib was the only independent predictive factor of developing ≥ grade 2 HFS (HR, 0.414; P = 0.004). Longer progression-free survival (PFS) was also observed in the SoraCele group (P = 0.039), although overall survival was not prolonged (P = 0.305). These results suggest that sorafenib + Celecoxib administration alleviated sorafenib-related skin toxicity. Longer PFS was achieved in clinical practice, although overall survival was not prolonged (ClinicalTrials.gov: NCT02961998).

10.
Mol Cell Endocrinol ; 494: 110492, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31255731

ABSTRACT

Approximately 20% of Graves' disease (GD) patients may result eventually in hypothyroidism in their natural course. Uterus globulin-associated protein 1 (UGRP1) was associated with GD in our previous study. Here we investigated the role of UGRP1 in the development of autoimmune thyroid disease (AITD). The results showed that UGRP1 was expressed in the thyrocytes of most Hashimoto's thyroiditis (HT) patients and a proportion of GD patients (293 HT and 198 GD). The pathologic features of UGRP1-positive thyrocytes resembled "Hürthle cells", and were surrounded by infiltrated leukocytes. The positivity rate of TPOAb in UGRP1-positive GD patients was much higher than that in -negative GD patients. Moreover, UGRP1 was co-expressed with Fas and HLA-DR in the thyrocytes of AITD patients. We also found IL-1ß but not Th1 or Th2 cytokines was able to upregulate the expression of UGRP1. Our findings indicated that UGRP1 may be a novel marker in thyrocytes to predict GD patients who develop hypothyroidism.


Subject(s)
Disease Progression , Graves Disease/metabolism , Graves Disease/pathology , Hypothyroidism/metabolism , Hypothyroidism/pathology , Secretoglobins/metabolism , Biomarkers/metabolism , HLA-DR Antigens/metabolism , Humans , Interleukin-1beta/metabolism , Secretoglobins/genetics , Thyroid Epithelial Cells/metabolism , Thyroid Gland/metabolism , Thyroid Gland/pathology , Up-Regulation/genetics , fas Receptor/metabolism
11.
Curr Med Sci ; 39(4): 582-588, 2019 Aug.
Article in English | MEDLINE | ID: mdl-31346994

ABSTRACT

Type 2 diabetes mellitus (T2DM) has become a prevalent health problem in China, especially in urban areas. Early prevention strategies are needed to reduce the associated mortality and morbidity. We applied the combination of rules and different machine learning techniques to assess the risk of development of T2DM in an urban Chinese adult population. A retrospective analysis was performed on 8000 people with non-diabetes and 3845 people with T2DM in Nanjing. Multilayer Perceptron (MLP), AdaBoost (AD), Trees Random Forest (TRF), Support Vector Machine (SVM), and Gradient Tree Boosting (GTB) machine learning techniques with 10 cross validation methods were used with the proposed model for the prediction of the risk of development of T2DM. The performance of these models was evaluated with accuracy, precision, sensitivity, specificity, and area under receiver operating characteristic (ROC) curve (AUC). After comparison, the prediction accuracy of the different five machine models was 0.87, 0.86, 0.86, 0.86 and 0.86 respectively. The combination model using the same voting weight of each component was built on T2DM, which was performed better than individual models. The findings indicate that, combining machine learning models could provide an accurate assessment model for T2DM risk prediction.


Subject(s)
Diabetes Mellitus, Type 2/epidemiology , Machine Learning , Risk Assessment , Adult , China/epidemiology , Cross-Sectional Studies , Diabetes Mellitus, Type 2/diagnosis , Diabetes Mellitus, Type 2/pathology , Female , Humans , Male , Retrospective Studies
12.
Clin Cancer Res ; 25(14): 4567-4579, 2019 07 15.
Article in English | MEDLINE | ID: mdl-30979744

ABSTRACT

PURPOSE: Neoadjuvant chemoradiotherapy (neoCRT) is a standard treatment for locally advanced rectal cancer (LARC); however, resistance to chemoradiotherapy is one of the main obstacles to improving treatment outcomes. The goal of this study was to identify factors involved in the radioresistance of colorectal cancer and to clarify the underlying mechanisms. EXPERIMENTAL DESIGN: A genome-wide RNAi screen was used to search for candidate radioresistance genes. After RFC4 knockdown or overexpression, colorectal cancer cells exposed to X-rays both in vitro and in a mouse model were assayed for DNA damage, cytotoxicity, and apoptosis. Moreover, the regulatory effects and mechanisms of RFC4 in DNA repair were investigated in vitro. Finally, the relationships between RFC4 expression and clinical parameters and outcomes were investigated in 145 patients with LARC receiving neoCRT. RESULTS: RFC4, NCAPH, SYNE3, LDLRAD2, NHP2, and FICD were identified as potential candidate radioresistance genes. RFC4 protected colorectal cancer cells from X-ray-induced DNA damage and apoptosis in vitro and in vivo. Mechanistically, RFC4 promoted nonhomologous end joining (NHEJ)-mediated DNA repair by interacting with Ku70/Ku80 but did not affect homologous recombination-mediated repair. Higher RFC4 expression in cancer tissue was associated with weaker tumor regression and poorer prognosis in patients with LARC treated with neoCRT, which likely resulted from the effect of RFC4 on radioresistance, not chemoresistance. CONCLUSIONS: RFC4 was identified as a radioresistance factor that promotes NHEJ-mediated DNA repair in colorectal cancer cells. In addition, the expression level of RFC4 predicted radiotherapy responsiveness and the outcome of neoadjuvant radiotherapy in patients with LARC.


Subject(s)
Colorectal Neoplasms/pathology , DNA End-Joining Repair , DNA Repair , Gene Expression Regulation, Neoplastic , RNA, Small Interfering/genetics , Radiation Tolerance/genetics , Replication Protein C/genetics , Animals , Apoptosis , Biomarkers, Tumor/genetics , Cell Proliferation , Chemoradiotherapy, Adjuvant , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Female , Genome, Human , High-Throughput Screening Assays , Humans , Ku Autoantigen/genetics , Ku Autoantigen/metabolism , Male , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Neoadjuvant Therapy , Prognosis , RNA Interference , Replication Protein C/antagonists & inhibitors , Survival Rate , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
13.
Aging (Albany NY) ; 10(10): 2755-2771, 2018 10 19.
Article in English | MEDLINE | ID: mdl-30341253

ABSTRACT

Chemoradiotherapy combined with surgical resection is the standard treatment for locally advanced rectal cancer, but not all the patients respond to neoadjuvant treatment. Transforming acidic coiled-coil protein-3 (TACC3) is frequently aberrantly expressed in rectal cancer tissue. In this study, we investigated whether TACC3 could serve as a biomarker predictive of the efficacy of chemoradiotherapy. In all, 152 rectal cancer patients with tumor tissue collected at biopsy and set aside before treatment were enrolled in this study. All patients received chemoradiotherapy and surgical resection. Immunohistochemically detected tumoral TACC3 expression significantly decreased sensitivity to chemoradiotherapy [risk ratio (RR) = 2.236, 95% confidence interval (CI): 1.447-3.456; P = 0.001] and thus the pathological complete response rate (P = 0.001). TACC3 knockdown using specific siRNA enhanced radiotherapy-induced decreases in proliferation and colony formation by HCT116 and SW480 cells and increased the incidence of radiotherapy-induced apoptosis. Cox multivariate analysis showed that TACC3 was a significant prognostic factor for overall survival (P = 0.017) and disease-free survival (P = 0.020). These findings suggest TACC3 expression may be predictive of chemoradiotherapy sensitivity and prognosis in locally advanced rectal cancer.


Subject(s)
Biomarkers, Tumor/metabolism , Chemoradiotherapy, Adjuvant , Digestive System Surgical Procedures , Microtubule-Associated Proteins/metabolism , Neoadjuvant Therapy , Rectal Neoplasms/therapy , Adult , Aged , Apoptosis/drug effects , Apoptosis/radiation effects , Biomarkers, Tumor/genetics , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Chemoradiotherapy, Adjuvant/adverse effects , Digestive System Surgical Procedures/adverse effects , Disease-Free Survival , Dose-Response Relationship, Drug , Female , HCT116 Cells , Humans , Male , Microtubule-Associated Proteins/genetics , Middle Aged , Neoadjuvant Therapy/adverse effects , Radiation Tolerance , Rectal Neoplasms/genetics , Rectal Neoplasms/metabolism , Rectal Neoplasms/pathology , Retrospective Studies , Time Factors
14.
World J Gastroenterol ; 24(20): 2181-2190, 2018 May 28.
Article in English | MEDLINE | ID: mdl-29853736

ABSTRACT

AIM: To evaluate indoleamine-2,3-dioxygenase 1/cyclooxygenase 2 (IDO1/COX2) expression as an independent prognostic biomarker for colorectal cancer (CRC) patients. METHODS: We retrospectively studied the medical records of 95 patients who received surgical resection from August 2008 to January 2010. All patients were randomly assigned to adjuvant treatment with or without celecoxib groups after surgery. We performed standard immunohistochemistry to assess the expression levels of IDO1/COX2 and evaluated the correlation of IDO1/COX2 with clinicopathological factors and overall survival (OS) outcomes. RESULTS: The expression of nuclear IDO1 was significantly correlated with body mass index (P < 0.001), and IDO1 expression displayed no association with sex, age, tumor differentiation, T stage, N stage, carcinoembryonic antigen, cancer antigen 19-9, CD3+ and CD8+ tumor infiltrating lymphocytes, and COX2. In univariate analysis, we found that nuclear IDO1 (P = 0.039), nuclear/cytoplasmic IDO1 [hazard ratio (HR) = 2.044, 95% confidence interval (CI): 0.871-4.798, P = 0.039], nuclear IDO1/COX2 (HR = 3.048, 95%CI: 0.868-10.7, P = 0.0049) and cytoplasmic IDO1/COX2 (HR = 2.109, 95%CI: 0.976-4.558, P = 0.022) all yielded significantly poor OS outcomes. Nuclear IDO1 (P = 0.041), nuclear/cytoplasmic IDO1 (HR = 3.023, 95%CI: 0.585-15.61, P = 0.041) and cytoplasmic IDO1/COX2 (HR = 2.740, 95%CI: 0.764-9.831, P = 0.038) have significantly poor OS outcomes for the CRC celecoxib subgroup. In our multivariate Cox model, high coexpression of cytoplasmic IDO1/COX2 was found to be an independent predictor of poor outcome in CRC (HR = 2.218, 95%CI: 1.011-4.48, P = 0.047) and celecoxib subgroup patients (HR = 3.210, 95%CI: 1.074-9.590, P = 0.037). CONCLUSION: Our results showed that cytoplasmic IDO1/COX2 coexpression could be used as an independent poor predictor for OS in CRC.


Subject(s)
Biomarkers, Tumor/metabolism , Colorectal Neoplasms/pathology , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/metabolism , Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism , Celecoxib/therapeutic use , Cell Nucleus/metabolism , Chemotherapy, Adjuvant/methods , China/epidemiology , Colorectal Neoplasms/mortality , Colorectal Neoplasms/therapy , Cytoplasm/metabolism , Epithelial Cells/pathology , Female , Follow-Up Studies , Humans , Intestinal Mucosa/cytology , Intestinal Mucosa/pathology , Lymphocytes, Tumor-Infiltrating , Male , Middle Aged , Prognosis , Retrospective Studies , Survival Analysis
15.
Saudi J Biol Sci ; 25(2): 372-376, 2018 Feb.
Article in English | MEDLINE | ID: mdl-29472793

ABSTRACT

The present study was aimed to investigate the effect of dihydroartemisinin on the colon cancer cell proliferation and apoptosis. The results from MTT assay revealed a concentration and time dependent relation between the inhibition of SW 948 cell viability and dihydroartemisinin addition. The viability of SW 948 cells was reduced to 45 and 24% on treatment with 30 and 50 µM, respectively concentrations of dihydroartemisinin after 48 h. Morphological examination of SW 948 cells showed attainment of rounded shape and cluster formation on treatment with dihydroartemisinin. Western blot analysis showed a significant increase in the activation of caspase-3 and expression of cleaved PARP by dihydroartemisinin treatment. The activation of PPARγ was increased significantly in SW 948 cells by treatment with dihydroartemisinin. Compared to control, the migration potential of SW 948 cells was reduced significantly (p < 0.005) and the expression levels of MMP-2 and -9 inhibited by dihydroartemisinin at 50 µM concentration. In the dihydroartemisinin treatment group colon tumor formation was significantly inhibited on treatment with 20 mg/kg doses of dihydroartemisinin after 30 days. Therefore, dihydroartemisinin inhibits colon cancer growth by inducing apoptosis and increasing the expression of PPARγ. Thus dihydroartemisinin can be used for the treatment of colon cancer.

16.
Sci Rep ; 7(1): 16043, 2017 11 22.
Article in English | MEDLINE | ID: mdl-29167471

ABSTRACT

Approximately 30% of locally advanced rectal cancer patients might not benefit from chemoradiotherapy; however, the response to neoadjuvant chemoradiotherapy in these cases is difficult to predict. Pim-3 is a member of the provirus integration site for a moloney murine leukemia virus family of proteins that contributes to cell proliferation, survival, and chemotherapy resistance. Therefore, the relationship between Pim-3 expression and response to neoadjuvant chemoradiotherapy in rectal cancer patients is important to evaluate. 175 rectal cancer patients who underwent neoadjuvant treatment enrolled in this study. The relationship between Pim-3 expression on immunohistochemical analysis of rectal cancer tissue, which was obtained before treatment, the response to chemoradiotherapy and survival was investigated. The patients with no Pim-3 expression were more likely to achieve a pathologic complete response to chemoradiotherapy than patients with Pim-3 expression (P = 0.001). Cox multivariate analysis showed that the significant prognostic factors were Pim-3 expression (P = 0.003) and the number of neoadjuvant chemotherapy cycles (P = 0.005) for overall survival. Neoadjuvant chemotherapy cycles (P = 0.007), adjuvant chemotherapy cycles (P = 0.004) and pathology types (P = 0.049) were significant prognostic factors for disease-free survival. Pim-3 is a potential predictive biomarker for the response of rectal cancer to chemoradiotherapy.


Subject(s)
Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins/metabolism , Rectal Neoplasms/drug therapy , Rectal Neoplasms/therapy , Adult , Capecitabine/therapeutic use , Chemoradiotherapy , Chemotherapy, Adjuvant , Female , Humans , Male , Middle Aged , Neoadjuvant Therapy , Oxaliplatin/therapeutic use , Rectal Neoplasms/metabolism , Rectal Neoplasms/pathology , Treatment Outcome
17.
Mol Med Rep ; 16(5): 5769-5778, 2017 Nov.
Article in English | MEDLINE | ID: mdl-28849238

ABSTRACT

The majority of colorectal cancers (CRCs) are hormone­dependent. Thus, endocrine therapy has become an attractive strategy to treat CRC. The aim of the present study was to investigate the inhibitory effect of combined tamoxifen (TAM) plus ß­estradiol (E2) treatment on human DLD­1 CRC cells. The human DLD­1 CRC cell line was treated with different concentrations of TAM, ß­estradiol, or a combination of these two agents. Cell viability was assessed using an MTT assay, while apoptosis was detected using flow cytometry analysis. Alterations in the RNA and protein levels of the apoptosis­associated factors cyclin D1 and survivin were measured in the treated DLD­1 cells using semi­quantitative polymerase chain reaction (sqPCR) and western blot analyses. Alterations in cellular migration ability were monitored using a Transwell migration assay. Treatment with TAM, ß­estradiol and TAM plus ß­estradiol inhibited DLD­1 cell viability. The flow cytometry results revealed that these drugs promoted cell apoptosis, and the Transwell migration assay results indicated that the reduction in cell migration was greater in the TAM+E2 treatment group when compared with each treatment alone. sqPCR and western blot analysis results demonstrated that TAM, E2 and a combination of the two affected survivin expression based on the drug concentration and the treatment duration; however, they demonstrated no significant effect on cyclin D1 expression. In conclusion, treatment of DLD­1 cells with TAM, ß­estradiol, or a combination of these two drugs, inhibited cell viability and migration, promoted cell apoptosis, and reduced the mRNA and protein expression levels of survivin in a dose­ and time­dependent manner. These results provide novel experimental basis for hormonal adjuvant therapy for the treatment of CRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Estradiol/administration & dosage , Inhibitor of Apoptosis Proteins/genetics , Tamoxifen/administration & dosage , Antineoplastic Agents, Hormonal/administration & dosage , Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Flow Cytometry , Gene Expression Regulation, Neoplastic/drug effects , Humans , Signal Transduction/drug effects , Survivin
18.
J Cancer Res Clin Oncol ; 143(12): 2581-2593, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28849265

ABSTRACT

PURPOSE: The safety and efficacy of intraoperative chemotherapy in colorectal cancer have not yet been extensively investigated. This randomized control trial was designed to compare the safety and efficacy of intraoperative chemotherapy in combination with surgical resection to those of traditional surgical resection alone. METHODS: From January 2011 to January 2016, 696 colorectal cancer patients were enrolled in this study: 341 patients were randomly assigned to the intraoperative chemotherapy, which consist of portal vein chemotherapy, intraluminal chemotherapy and intraperitoneal chemotherapy, plus surgery group, whereas 344 patients were randomized to the control group to undergo surgery alone. Eleven patients withdrew consent. RESULTS: Intraoperative chemotherapy did not increase the rate of surgical complications, and no severe chemotherapy-associated side effects were observed. Four patients in each of the intraoperative chemotherapy and the control groups experienced anastomotic leakage and underwent a second operation (1.2 vs. 1.2%, P = 0.99). There were no deaths within 90 days after surgery in the chemotherapy group, whereas one patient died in the control group. Intraoperative chemotherapy did not decrease the rate of patients who received postoperative chemotherapy between the intraoperative group and control group (29.3 vs. 30.2%, P = 0.795). CONCLUSIONS: Intraoperative chemotherapy can be safely performed during colorectal surgery; however, follow-up is necessary for a better assessment of its efficacy. ClinicalTrial.gov Register Number: NCT01465451.


Subject(s)
Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/surgery , Fluorouracil/administration & dosage , Fluorouracil/adverse effects , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Humans , Intraoperative Care/methods , Intraoperative Complications/etiology , Middle Aged , Postoperative Complications/etiology , Prospective Studies
19.
World J Surg Oncol ; 15(1): 138, 2017 Jul 27.
Article in English | MEDLINE | ID: mdl-28750680

ABSTRACT

BACKGROUND: It is still under debate that whether stage IV colorectal cancer patients with unresectable metastasis can benefit from primary tumor resection, especially for asymptomatic colorectal cancer patients. Retrospective studies have shown controversial results concerning the benefit from surgery. This retrospective study aims to evaluate whether the site of primary tumor is a predictor of palliative resection in asymptomatic stage IV colorectal cancer patients. METHODS: One hundred ninety-four patients with unresectable metastatic colorectal cancer were selected from Sun Yat-sen University Cancer Center Database in the period between January 2007 and December 2013. All information was carefully reviewed and collected, including the treatment, age, sex, carcinoembryonic antigen, site of tumor, histology, cancer antigen 199, number of liver metastases, and largest diameter of liver metastasis. The univariate and multivariate analyses were used to detect the relationship between primary tumor resection and overall survival of unresectable stage IV colorectal cancer patients. RESULTS: One hundred twenty-five received palliative resection, and 69 received only chemotherapy. Multivariate analysis indicated that primary tumor site was one of the independent factors (RR 0.569, P = 0.007) that influenced overall survival. For left-side colon cancer patients, primary tumor resection prolonged the median overall survival time for 8 months (palliative resection vs. no palliative resection: 22 vs. 14 months, P = 0.009); however, for right-side colon cancer patients, palliative resection showed no benefit (12 vs. 10 months, P = 0.910). CONCLUSIONS: This study showed that left-side colon cancer patients might benefit from the primary tumor resection in terms of overall survival. This result should be further explored in a prospective study.


Subject(s)
Colorectal Neoplasms/mortality , Colorectal Neoplasms/surgery , Liver Neoplasms/pathology , Palliative Care/methods , Adult , Aged , Antigens, Tumor-Associated, Carbohydrate/blood , Asymptomatic Diseases/mortality , Carcinoembryonic Antigen/blood , China/epidemiology , Colorectal Neoplasms/blood , Colorectal Neoplasms/pathology , Female , Humans , Kaplan-Meier Estimate , Liver Neoplasms/secondary , Male , Middle Aged , Multivariate Analysis , Neoplasm Staging , Prognosis , Retrospective Studies , Young Adult
20.
Chin J Cancer ; 36(1): 31, 2017 Mar 20.
Article in English | MEDLINE | ID: mdl-28320486

ABSTRACT

BACKGROUND: Laparoscopic hepatectomy for hepatocellular carcinoma (HCC) located in segment VI, VII, or VIII of the liver is usually difficult because of poor operative exposure, due to the unique anatomical structure. In this study, we evaluated the practice of laparoscopic hepatectomy with the left jackknife position for patients with HCC located in segment VI, VII, or VIII. METHODS: A total of 10 patients were enrolled to undergo laparoscopic hepatectomy with the left jackknife position. Tumors located in segment VI, VII, or VIII were assessed by preoperative dynamic computed tomography or magnetic resonance imaging. Operation time, intraoperative blood loss, postoperative fasting time, postoperative drainage time, major postoperative complications, and duration of postoperative hospital stay were recorded. RESULTS: All surgeries were successfully completed. None of the patients required conversion to open surgery during the procedure, and no serious postoperative complications were observed. The median tumor size was 31 mm (range 23-41 mm) in diameter, the mean operation time was 166 ± 38 min, the mean intraoperative blood loss was 220 ± 135 mL, and the median postoperative hospital stay was 4 days (range 2-7 days). CONCLUSIONS: For HCC located in segment VI, VII, or VIII, laparoscopic hepatectomy with this novel position-the left jackknife position-is safe and effective during tumor resection by exposing a sufficient operating field. Trial registration ClinicalTrials.gov ID: NCT02809287.


Subject(s)
Carcinoma, Hepatocellular/surgery , Hepatectomy/methods , Laparoscopy/methods , Liver Neoplasms/surgery , Posture , Adult , Aged , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...