Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Int Immunopharmacol ; 131: 111887, 2024 Apr 20.
Article in English | MEDLINE | ID: mdl-38503018

ABSTRACT

BACKGROUND: The purpose of this study was to explore the dynamic changes of genomic mutations and their correlations with the efficacy in metastatic colorectal cancer (mCRC) patients treated with cetuximab plus mFOLFOX as the first-line treatment. METHODS: We included mCRC patients from January 2018 to October 2020 as a studied cohort which were treated with cetuximab plus mFOLFOX as first line therapy. Blood samples were collected for circulating tumor DNA (ctDNA) test at three timepoints: before the first-line therapy(baseline), at the time of first-line progression and at the time of second-line progression. Progression-free survival was considered as the primary endpoint while objective response rate and overall survival were determined as the secondary endpoints. RESULTS: Totally 39 patients received first-line treatment, of which 25 patients entered the second-line treatment, while 10 patients entered the third-line treatment. The median follow-up time was 16.4 months (95 %CI, 14.8-19.3). Along the treatment from first-line progress disease (PD) to second-line PD, proportions of TP53 (12/18, 67 %), APC (10/18, 56 %), FBXW7 (3/18, 17 %), and AMER1 (2/18, 11 %) were gradually increased according to results of single nucleotide variation (SNV). CONCLUSIONS: Resistant gene mutations caused by anti-EGFR drugs in RAS/BRAF wild-type mCRC patients can be observed by dynamic ctDNA analysis. TP53 and AMER1 mutations, tumor mutational burden (TMB) levels, and TP53/AMER1 co-mutation may predict the efficacy of the first-line cetuximab-contained treatment. Situations of genetic mutations were differentiated from first-line PD to second-line PD, which indicated that mutation detection may contribute to predict prognosis of mCRC patients.


Subject(s)
Colonic Neoplasms , Colorectal Neoplasms , Rectal Neoplasms , Humans , Cetuximab/therapeutic use , Proto-Oncogene Proteins B-raf/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Colonic Neoplasms/drug therapy , Rectal Neoplasms/drug therapy , Mutation , Antineoplastic Combined Chemotherapy Protocols/adverse effects
2.
Front Pharmacol ; 14: 1190967, 2023.
Article in English | MEDLINE | ID: mdl-37900166

ABSTRACT

Background: Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) is a highly aggressive malignancy with a poor prognosis. However, there are no consensus treatment guidelines, and decisions are usually extrapolated from intrahepatic cholangiocarcinoma (ICC) or hepatocellular carcinoma (HCC). Given that cHCC-CCA owns the unequivocal presence of both hepatocytic and cholangiocytic differentiation, a combination regimen of anti-PD1 antibody, multikinase inhibitor, and chemotherapy targeting against both components might be an optimal choice. Case presentation: We present the case of a patient with postoperative metastatic chemotherapy-resistant cHCC-CCA who exhibited a durable response and reasonable tolerability to a combination therapy consisting of the anti-PD1 antibody sintilimab, multikinase inhibitor lenvatinib, and nab-paclitaxel, despite having a low tumor mutational burden (TMB-L), microsatellite stability (MSS), and negative programmed cell death 1 ligand 1 (PD-L1). Conclusion: The combination regimen of immune checkpoint inhibitor sintilimab, multikinase inhibitor lenvatinib, and chemotherapy with nab-paclitaxel, which targets both the HCC and ICC components, may represent a promising treatment option for patients with cHCC-CCA. Further research is warranted to validate these findings in larger patient cohorts.

3.
Eur J Surg Oncol ; 48(7): 1559-1566, 2022 07.
Article in English | MEDLINE | ID: mdl-35115213

ABSTRACT

PURPOSE: The aim of the study was to comprehensively understand the combined hepatocellular and cholangiocarcinoma (CHC) and develop a nomogram for prognostic prediction of CHC. METHODS: Data were collected from the Surveillance, Epidemiology and End Results (SEER) database (year 2004-2014). Propensity-score matching (PSM) was used to match the demographic characteristic of the CHC versus hepatocellular carcinoma (HCC) and intrahepatic cholangiocarcinoma (ICC). A nomogram model was established to predict the prognosis in terms of cancer specific survival (CSS). The established nomogram was externally validated by a multicenter cohort. RESULTS: A total of 71,756 patients enrolled in our study including 62,877 HCC patients, 566 CHC patients, and 8303 ICC patients. The CHC, HCC, and ICC are not exactly similar in clinical characteristic. After PSM, the CSS of CHC was better than HCC but comparable to ICC. Tumor size, M stage, surgery, chemotherapy, and surgery were independently prognostic factors of CHC and were included in the establishment of novel nomogram. The c-index of the novel nomogram in SEER training set and multicenter validation was 0.779 and 0.780, respectively, which indicated that the model was with better discrimination power. In addition, decision curve analyses proved the favorable potential clinical effect of the predictive model. Lastly, a risk classification based on nomogram also verified the reliability of the model. CONCLUSION: CHC had better survival than HCC but was comparable to ICC. The nomogram was established based on tumor size, M stage, chemotherapy, surgery, and radiotherapy and well validated by external multicenter cohort.


Subject(s)
Bile Duct Neoplasms , Carcinoma, Hepatocellular , Cholangiocarcinoma , Liver Neoplasms , Bile Duct Neoplasms/pathology , Bile Ducts, Intrahepatic/pathology , Carcinoma, Hepatocellular/pathology , Cholangiocarcinoma/pathology , Humans , Liver Neoplasms/pathology , Nomograms , Prognosis , Reproducibility of Results , SEER Program
4.
Cancer Med ; 10(10): 3388-3402, 2021 05.
Article in English | MEDLINE | ID: mdl-33939281

ABSTRACT

BACKGROUND: To compare the efficacy of first-line bevacizumab plus chemotherapy with cetuximab plus chemotherapy based on the stratification of metastatic colorectal cancer (mCRC) patients with mucinous adenocarcinoma (MA) or mucinous component (MC). METHODS: A retrospective study involving all mCRC patients receiving first-line bevacizumab-based or cetuximab-based chemotherapy at our hospital from September 2013 to January 2020 was conducted. Overall survival (OS), progression-free survival (PFS), and objective response rate (ORR) were compared between the cetuximab-chemotherapy group and the bevacizumab-chemotherapy group on the basis of the conventional pathological classification of MA or MC. RESULTS: A total of 620 patients with mCRC were included in our study, consisting of 141 (22.7%) patients with MA/MC and 479 (77.3%) patients with non-mucinous adenocarcinoma (NMA). In the MA/MC cohort, patients who were treated with bevacizumab-based chemotherapy were associated with significantly better OS than those treated with cetuximab-base chemotherapy (30.0 vs. 26.3 months, p = 0.002), irrespective of tumor sites. The efficacy of bevacizumab-based chemotherapy was higher in nearly all subgroups as shown in the subgroup analysis. In the NMA cohort, median OS was better in the cetuximab plus chemotherapy group than that in the bevacizumab plus chemotherapy group (32.2 vs. 27.0 months, p = 0.005) for left-side mCRC patients, whereas OS was significantly longer in the bevacizumab plus chemotherapy group for right-side mCRC patients (26.0 vs. 20.9 months, p = 0.013). CONCLUSION: Conventional pathological classification (e.g. MA/MC) should be considered when tailoring the individualized optimal treatment for mCRC. Bevacizumab plus chemotherapy as first-line therapy may be the optimal option for patients with MA/MC.


Subject(s)
Adenocarcinoma, Mucinous/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Cetuximab/therapeutic use , Colorectal Neoplasms/drug therapy , Female , Humans , Male , Middle Aged , Progression-Free Survival , Retrospective Studies
5.
Oncologist ; 26(8): e1320-e1326, 2021 08.
Article in English | MEDLINE | ID: mdl-33830591

ABSTRACT

LESSONS LEARNED: Bevacizumab combined with S-1 and raltitrexed demonstrated positive antitumor efficacy and acceptable toxicity. This combination might represent a treatment option for refractory metastatic colorectal cancer. BACKGROUND: In patients with metastatic colorectal cancer (mCRC) refractory to standard therapies, S-1 plus raltitrexed showed a good objective response rate (ORR) and significant survival benefit in our previous study. In the present study, we assessed the activity and safety of bevacizumab combined with S-1 and raltitrexed. METHODS: This investigator-initiated, open-label, single-arm, phase II trial was performed at West China Hospital in China. Patients with mCRC who had disease progression after fluoropyrimidine, irinotecan, and oxaliplatin and had at least one measurable lesion were eligible for this trial. Anti-epidermal growth factor receptor (EGFR) (for tumors with wild-type RAS) and anti-vascular endothelial growth factor (VEGF) therapy in the first or second line was allowed, but patients who had been treated with bevacizumab across two consecutive chemotherapy regimens were excluded. Patients received bevacizumab (7.5 mg/kg on day 1), oral S-1 (80-120 mg per day for 14 days), and raltitrexed (3 mg/m2 on day 1) every 3 weeks. The primary endpoint was ORR. Secondary endpoints included progression-free survival (PFS), overall survival (OS), and toxicity. RESULTS: From September 2015 to November 2019, 44 patients were enrolled. Tumor response evaluation was available in 44 patients at the time of the analysis. There were no complete responses; the ORR was 15.9%, and the disease control rate was 54.5%. Median PFS and OS were 110 days (95% confidence interval [CI], 65.0-155.0) and 367 days (95% CI, 310.4-423.6), respectively. The combination was well tolerated. CONCLUSION: Bevacizumab combined with S-1 and raltitrexed showed promising antitumor activity and safety in refractory mCRC.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Bevacizumab/therapeutic use , Colorectal Neoplasms/drug therapy , Fluorouracil/therapeutic use , Humans , Leucovorin/therapeutic use , Quinazolines/therapeutic use , Thiophenes
6.
Front Oncol ; 10: 570268, 2020.
Article in English | MEDLINE | ID: mdl-33324548

ABSTRACT

BACKGROUND: Signet ring cell containing gastric cancer (SRCGC) is a rare subtype of gastric cancer, and its adjuvant therapy is based on general gastric cancer. However, the effectiveness of radiotherapy for those SRCGC patients remains unknown. PURPOSE: The purpose of the study was to analyze whether the addition of radiotherapy to adjuvant chemotherapy (CT) can benefit survival in resected SRCGC patients. METHODS: Patients with SRCGC, who underwent D2 gastrectomy followed by adjuvant chemotherapy or chemoradiotherapy (CRT), were retrospectively collected. According to the proportion of signet ring cells, patients were histologically classified as pure SRCGC (pSRCGC) containing 100% of signet ring cells, mixed SRCGC (mSRCGC) containing >50% of signet ring cells, and contaminated SRCGC (cSRCGC) containing <50% of signet ring cells. Among the 272 patients, 156 were treated by CT alone and 116 by CRT. The primary endpoint was 3-year overall survival rate (3-year OS rate). RESULTS: With a median follow-up of 80.5 months, the 3-year OS rate was significantly higher in the CT group (70.5% vs. 58.6%, HR = 0.633, P = 0.017) compared with CRT group. Three independent characteristics were predictive of a poor overall survival: CRT treatment (P = 0.019), tumor size ≥5 cm (P < 0.001), and the presence of vessel invasion (P = 0.009). Subgroup analyses showed CRT significantly impaired prognosis in SRCGC patients in the cSRCGC subset, as well as lesions located in lower-middle sites, subtotal gastrectomy, male, <60 year, and no vessel invasion. Peritoneal was the most common recurrence site in SRCGC patients. The adverse events leukopenia and neutropenia were more common in the CRT group (P = 0.007). CONCLUSIONS: Adjuvant chemoradiotherapy was associated with poor survival compared with adjuvant chemotherapy in SRCGC patients with D2 gastrectomy.

7.
FASEB J ; 34(8): 11185-11199, 2020 08.
Article in English | MEDLINE | ID: mdl-32645243

ABSTRACT

The efficacy of chimeric antigen receptor T (CAR-T) cell therapy in solid tumors is far from satisfactory. In this study, we investigated the influence of epithelial-mesenchymal transition (EMT) on the antitumor effect of CAR-T cells and explored the potential efficacy of combining CAR-T cells with inhibitors targeting EMT. We successfully induced EMT in tumor cells with TGF-ß1, and the antitumor effect of HER2-directed CAR-T cells was significantly suppressed by EMT. Upregulation of PD-L1 was observed in tumor cells undergoing EMT, and change in PD-L1 expression during the EMT process was dependent on the MEK/ERK and PI3K/Akt pathways. Inhibition of the TGF-ß1 pathway could block the EMT process in tumor cells and restore their susceptibility to HER2-directed CAR-T cells in vitro. In addition, targeting the TGF-ß1 pathway significantly enhanced the antitumor effect of HER2-directed CAR-T cells in vivo. Our findings suggest that blocking EMT could potently enhance the antitumor effect of CAR-T cells, which provides a promising approach to improving the therapeutic efficacy of CAR-T cell therapy in solid tumors.


Subject(s)
Antineoplastic Agents/immunology , Epithelial-Mesenchymal Transition/immunology , Neoplasms/immunology , Neoplasms/therapy , Receptor, ErbB-2/immunology , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , A549 Cells , Animals , B7-H1 Antigen/immunology , Cell Line , Cell Line, Tumor , Female , HCT116 Cells , HEK293 Cells , HT29 Cells , Humans , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred NOD , Mice, SCID , Signal Transduction/immunology , Transforming Growth Factor beta1/immunology , Xenograft Model Antitumor Assays/methods
8.
Medicine (Baltimore) ; 99(14): e19413, 2020 Apr.
Article in English | MEDLINE | ID: mdl-32243360

ABSTRACT

The aim of this observational study was to test whether ABO blood type was a prognostic factor for pancreatic ductal adenocarcinoma (PDAC) patients and whether other risk factors could influence pancreatic cancer patients' survival. This study included 610 patients who were diagnosed as pancreatic cancer and had undergone radical surgery. Patients' characteristics included age, gender, tumor stage, tumor grade, adenosquamous carcinoma (ASC) status, preoperative serum carbohydrate antigen 19-9 (CA19-9) levels, preoperative serum carcinoembryonic antigen (CEA) levels, ABO blood type, smoking status, and drinking status were analyzed in this study. Cox proportional hazards regression model and Kaplan-Meier method were used to evaluate the role of prognostic factors. For pancreatic cancer patients undergoing radical surgery, the overall survival was worse for ASC patients than PDAC patients (Log-rank = 11.315, P < .001). Compared with ASC patients (Log-rank < 0.001, P = .996), PDAC patients can benefit from chemotherapy (Log-rank = 17.665, P < .001). For PDAC patients, O blood type had better overall survival than non-O blood type (Log-rank = 4.153, P = .042). Moreover, the group with higher serum levels of CA19-9 had poor prognosis compared to another group with low serum CA19-9 (Log-rank = 4.122, P = .042). Higher CEA levels indicated poor prognosis (Log-rank = 13.618, P < .001). In conclusion, ASC status was associated with overall survival of pancreatic cancer patients and cannot benefit from postoperative chemotherapy. Non-O blood type was a prognostic factor for PDAC patients.


Subject(s)
Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , ABO Blood-Group System/blood , Adult , Age Factors , Aged , Alcohol Drinking/epidemiology , CA-19-9 Antigen/blood , Carcinoembryonic Antigen/blood , Carcinoma, Adenosquamous/pathology , Carcinoma, Pancreatic Ductal/epidemiology , Carcinoma, Pancreatic Ductal/surgery , Cigarette Smoking/epidemiology , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/surgery , Proportional Hazards Models , Risk Factors , Sex Factors
9.
FASEB J ; 34(1): 1768-1782, 2020 01.
Article in English | MEDLINE | ID: mdl-31914650

ABSTRACT

Interleukin-18 (IL-18) has been demonstrated to augment the antitumor capacity of chimeric antigen receptor-T cells (CAR-T) but the underlying mechanisms are largely unknown. Here we explored the effects and mechanisms of exogenous IL-18 on the antitumor response of CAR-T cells. IL-18 boosted the cytotoxicity of human epidermal growth factor receptor-2 (HER2)-specific CAR-T cells ex vivo and enhanced the antitumor efficacy of the CAR-T cells in immunodeficient mice, moreover, IL-18 improved the antitumor capacity of OVA-specific T cells in immunocompetent mice, indicating the universal enhancing function of IL-18 for adoptive cell therapy. To address the roles of IL-18 receptor (IL-18R) in the enhancing function, we evaluated the effects of IL-18R knockout (IL-18R-/-) condition in immunocompetent host and CAR-T cells on the IL-18-enhanced antitumor activities. Interestingly, IL-18 persisted to improve the antitumor ability of IL-18R intact CAR-T cells in IL-18R-/- mice. For IL-18R-/- CAR-T cells, however, IL-18 still holds the enhancing ability to boost the antitumor efficacy in IL-18R-/- mice, albeit the ex vivo tumor-killing ability was lower than that of IL-18R intact CAR-T cells, indicating that IL-18R-independent pathway is involved in the enhancement. Furthermore, tagged IL-18 binded to the membrane of IL-18R-/- splenic and lymph node cells and IL-18R intact and IL-18R-/- CAR-T cells showed distinct transcriptomic profiles when stimulated by IL-18. These data demonstrate that IL-18R-independent pathways contribute to functions of IL-18.


Subject(s)
Antineoplastic Agents/metabolism , Interleukin-18/metabolism , Receptors, Antigen, T-Cell/metabolism , Receptors, Interleukin-18/metabolism , Signal Transduction/physiology , T-Lymphocytes/metabolism , Animals , Cell Line , Female , HEK293 Cells , Humans , Immunotherapy, Adoptive/methods , Mice , Mice, Inbred C57BL , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays/methods
10.
Cell Death Dis ; 10(10): 722, 2019 Sep 26.
Article in English | MEDLINE | ID: mdl-31558703

ABSTRACT

Since publication of this article, the authors have noticed that there were errors in Fig. 1b (the CT 26 cells colony formation images) and Fig. 7c (the vehicle group images). As a result of the misfiling of the data during preparation of figures, incorrect images were inadvertently inserted in these figures.An amendment to this paper has been published and can be accessed via a link at the top of the paper.

11.
Hum Gene Ther ; 30(4): 402-412, 2019 04.
Article in English | MEDLINE | ID: mdl-30693795

ABSTRACT

Chimeric antigen receptor-modified T cells (CAR-T cells) have emerged as a promising cancer immunotherapy for solid tumors. Epithelial cell adhesion molecule (EpCAM) is overexpressed in a variety of tumors and is recognized as a biomarker for circulating tumor cells and cancer stem cells, representing an attractive target for adoptive T-cell immunotherapy. This study generated third-generation CAR-T cells with redirected specificity to EpCAM (EpCAM CAR-T) by lentiviral vector. The study demonstrated that EpCAM CAR-T cells can elicit lytic cytotoxicity to target cells in an EpCAM-dependent manner and secrete cytotoxic cytokines, including interferon gamma and tumor necrosis factor alpha. Furthermore, adoptive transfer of EpCAM CAR-T cells significantly delayed tumor growth and formation in xenograft models. In addition, the safety evaluation showed that CAR-T cells have no systemic toxicity in mice. The data confirmed the antitumor ability and safety of CAR-T cells targeting EpCAM and may provide a new target for CAR-T cell therapies in treating solid tumors.


Subject(s)
Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Epithelial Cell Adhesion Molecule/immunology , Immunotherapy, Adoptive , Receptors, Antigen, T-Cell/metabolism , Receptors, Chimeric Antigen/metabolism , T-Lymphocytes/immunology , Animals , Biomarkers , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/therapy , Cytotoxicity, Immunologic , Disease Models, Animal , Drug Evaluation, Preclinical , Epithelial Cell Adhesion Molecule/antagonists & inhibitors , Humans , Immunophenotyping , Immunotherapy, Adoptive/adverse effects , Immunotherapy, Adoptive/methods , Mice , Receptors, Antigen, T-Cell/genetics , Receptors, Chimeric Antigen/genetics , T-Lymphocytes/metabolism , Xenograft Model Antitumor Assays
12.
Oncologist ; 24(5): 591-e165, 2019 05.
Article in English | MEDLINE | ID: mdl-30651398

ABSTRACT

LESSONS LEARNED: The upregulation of dihydropyrimidine dehydrogenase (DPD) and thymidylate synthase (TS) are important mechanisms of resistance to 5-fluorouracil (5-FU) in metastatic colorectal cancer (mCRC) after long exposure to 5-FU.S-1 (containing a DPD inhibitor) combined with raltitrexed (a TS inhibitor) showed a moderate effect, which needs further study as a third- or later-line therapy in mCRC. BACKGROUND: 5-fluorouracil (5-FU) is a fundamental drug in the treatment of metastatic colorectal cancer (mCRC). Patients with mCRC are often exposed to 5-FU and/or its analogues for a long time because of its central role in treatment regimens. The upregulation of dihydropyrimidine dehydrogenase (DPD) and/or thymidylate synthase (TS) are important mechanisms of resistance of 5-FU. To evaluate the efficacy and safety of S-1 (containing a DPD inhibitor) and raltitrexed (a TS inhibitor) for refractory mCRC, a one-center, single-arm, prospective phase II trial was conducted. METHODS: Patients who had mCRC that had progressed after treatment with fluoropyrimidine, irinotecan, and oxaliplatin and who had at least one measurable lesion were eligible for this trial. Patients received oral S-1 (80-120 mg for 14 days every 3 weeks) plus an intravenous infusion of raltitrexed (3 mg/m2 on day 1 every 3 weeks). The primary endpoint was objective response rate (ORR). Secondary endpoints included progression-free survival (PFS), overall survival (OS), and toxicity. RESULTS: In total, 46 patients were enrolled. Three patients did not complete the first assessment because of adverse events and unwillingness, leaving tumor response evaluation available in 43 patients. Of 43 evaluable patients, the ORR was 13.9% and disease control rate was 58.1%. In the intention-to-treat population (n = 46), the ORR was 13.0% and disease control rate was 54.3%. Median PFS and median OS were 107 days (95% confidence interval [CI], 96.3-117.7) and 373 days (95% CI, 226.2-519.8), respectively. Most of the adverse effects were mild to moderate. CONCLUSION: S-1 combined with raltitrexed for refractory mCRC showed moderate effect, and it is worthy of further study as third- or later-line therapy in mCRC.


Subject(s)
Colorectal Neoplasms/drug therapy , Oxonic Acid/therapeutic use , Quinazolines/therapeutic use , Tegafur/therapeutic use , Thiophenes/therapeutic use , Adult , Aged , Colorectal Neoplasms/pathology , Drug Combinations , Female , Humans , Male , Middle Aged , Neoplasm Metastasis , Oxonic Acid/pharmacology , Quinazolines/pharmacology , Tegafur/pharmacology , Thiophenes/pharmacology
13.
Gene Ther ; 25(8): 538-547, 2018 12.
Article in English | MEDLINE | ID: mdl-30254305

ABSTRACT

Mesenchymal stem cells (MSCs), well-studied adult stem cells in various tissues, possess multi-lineage differentiation potential and anti-inflammatory properties. MSCs have been approved to regenerate lineage-specific cells to replace injured cells in tissues. MSCs are approved to treat inflammatory diseases. With the discovery of genes important for the repair of damaged tissues, MSCs genetically modified by such genes hold improved therapeutic potential. In this review, we summarised the uses of genetically modified MSCs to treat different diseases, including bone diseases, cardiovascular diseases, autoimmune diseases, central nervous system disorders, and cancer. To better understand the exact role of genetically modified MSCs, key mechanisms determining, which genes are selected to be used for modifying MSCs and improvements in post-genetic modification are discussed. Therapeutic benefits enhanced by genetic modifications are to be documented by further clinical studies.


Subject(s)
Genetic Therapy/methods , Mesenchymal Stem Cell Transplantation/methods , Animals , Gene Transfer Techniques , Humans , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism
14.
Mol Med Rep ; 18(2): 2335-2341, 2018 Aug.
Article in English | MEDLINE | ID: mdl-29956798

ABSTRACT

Recent studies have demonstrated that some chemotherapeutic drugs can enhance antitumor immunity by eliminating and inactivating immunosuppressive cells. Oxaliplatin (OXP) induces immunogenic cell death by increasing the immunogenicity of cancer cells. However, the effects of OXP on the tumor immunosuppressive microenvironment remain unclear. The aim of the present study was to evaluate the antitumor activity of OXP by intraperitoneal (i.p.) administration in an abdominal implantation model of colon cancer and tested the tumor immune microenvironment to observe whether OXP affects the local immune inhibitory cell populations. Abdominal metastasis models were established by inoculation of CT26 cells. The antitumor efficacy of OXP and the tumor immune microenvironment were evaluated. The tumors and spleens of mice were harvested for flow cytometric analysis. Cluster of differentiation (CD)­8+CD69+ T cells, regulatory T cells (Tregs), CD11b+F4/80high macrophages and myeloid­derived suppressor cells (MDSCs) were evaluated by flow cytometric analysis. In vivo i.p. administration of OXP inhibited tumor growth in the abdominal metastasis model. Furthermore, OXP was observed to increase tumor­infiltrating activated CD8+ T cells in tumors, decrease CD11b+F4/80high macrophages in tumors and decrease MDSCs in the spleen. These results suggested that i.p. administration of OXP alone may inhibit tumor cell growth and induce the antitumor immunostimulatory microenvironment by eliminating immunosuppressive cells.


Subject(s)
Cell Proliferation/drug effects , Colonic Neoplasms/drug therapy , Organoplatinum Compounds/administration & dosage , Abdomen/pathology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Colonic Neoplasms/immunology , Colonic Neoplasms/pathology , Humans , Immunosuppression Therapy/methods , Injections, Intraperitoneal , Mice , Oxaliplatin , Spleen/immunology , Spleen/pathology , Spleen/transplantation , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
15.
Oncotarget ; 8(32): 52366-52380, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28881736

ABSTRACT

OBJECTIVES: The safety and surgical oncology of laparoscopy-assisted total gastrectomy (LATG) remain inconclusive and challenging. This study aimed to compare the short-term and long-term outcomes between LATG and open total gastrectomy (OTG) procedures. RESULTS: In the all-included analyses, there were 69 patients in the LATG group and 268 in the OTG group. LATG was as safe as OTG without increasing postoperative morbidity and mortality. Stage imbalance might introduce differences in the numbers of harvested lymph nodes in LATG (34.4 ± 12.0) and OTG (40.9 ± 16.9), whereas 95.7% of patients underwent D2/D2+ dissection during the LATG procedure. After a median 31 months of follow-up, the overall survival outcomes were comparable between the LATG and OTG procedures (HR = 1.16, 95% CI 0.68-1.97). Sensitivity analysis found comparable node retrieval and stage-specific or treatment-specific overall survival. MATERIALS AND METHODS: A retrospective case-control study was conducted among gastric cancer patients who underwent either LATG or OTG with curative intention between June 2006 and December 2015. Data retrieval was based on the Surgical Gastric Cancer Patient Registry in the West China Hospital. The primary outcome was overall survival. The secondary outcomes were postoperative complication incidence and severity, operation duration, blood loss, number of harvested lymph nodes, and postoperative hospital stay. Matched pairwise case-control comparisons were performed as a sensitivity analysis. CONCLUSIONS: LATG by experienced surgeons possibly has comparable short-term surgical outcomes and long-term survival outcomes compared with OTG for gastric cancer patients. However, high-quality RCTs are necessary before confirmative judgment and recommendation as an optional treatment in general practice.

16.
Cell Death Dis ; 8(1): e2534, 2017 01 05.
Article in English | MEDLINE | ID: mdl-28055016

ABSTRACT

Colorectal carcinoma (CRC) is the one of the most common cancers with considerable metastatic potential, explaining the need for new drug candidates that inhibit tumor metastasis. The signal transducers and activators of the transcription 3 (Stat3) signaling pathway has an important role in CRC and has been validated as a promising anticancer target for CRC therapy. In the present study, we report our findings on nifuroxazide, an antidiarrheal agent identified as an inhibitor of Stat3. Our studies showed that nifuroxazide decreased the viability of three CRC cell lines and induced apoptosis of cancer cells in a concentration-dependent manner. Moreover, western blot analysis demonstrated that the occurrence of its apoptosis was correlated with the activation of Bax and cleaved caspase-3, and decreased the expression of Bcl-2. In addition, nifuroxazide markedly impaired CRC cell migration and invasion by downregulating phosphorylated-Stat3Tyr705, and also impaired the expression of matrix metalloproteinases (MMP-2 and MMP-9). Furthermore, our studies showed that nifuroxazide also significantly inhibited the tumor metastasis in lung and abdomen metastasis models of colon cancer. Meanwhile, nifuroxazide functionally reduced the proliferation index, induced tumor apoptosis and impaired metastasis. Notably, nifuroxazide reduced the number of myeloid-derived suppressor cells in the blood, spleens and tumors, accompanied by the increased infiltration of CD8+ T cells in the tumors. Importantly, a marked decrease in the number of M2-type macrophages in tumor in the abdomen metastasis model was also observed. Taken together, our results indicated that nifuroxazide could effectively inhibit tumor metastasis by mediating Stat3 pathway and it might have a therapeutic potential for the treatment of CRC.


Subject(s)
Apoptosis/genetics , Colorectal Neoplasms/drug therapy , Neoplasm Metastasis/drug therapy , STAT3 Transcription Factor/genetics , Animals , Cell Line, Tumor , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Humans , Hydroxybenzoates/administration & dosage , Mice , Neoplasm Metastasis/genetics , Neoplasm Proteins/biosynthesis , Nitrofurans/administration & dosage , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
17.
Oncotarget ; 8(1): 1788-1795, 2017 Jan 03.
Article in English | MEDLINE | ID: mdl-27705946

ABSTRACT

OBJECTIVES: To compare the proportion of stage I lung cancer and population mortality in China to those in U.S. and Europe where lung cancer screening by low-dose computed tomography (LDCT) has been already well practiced. METHODS: The proportions of stage I lung cancer in LDCT screening population in U.S. and Europe were retrieved from NLST and NELSON trials. The general proportion of stage I lung cancer in China was retrieved from a rapid meta-analysis, based on a literature search in the China National Knowledge Infrastructure database. The lung cancer mortality and prevalence of China, U.S. and Europe was retrieved from Globocan 2012 fact sheet. Mortality-to-prevalence ratio (MPR) was applied to compare the population survival outcome of lung cancer. RESULTS: The estimated proportion of stage I lung cancer in China is merely 20.8% among hospital-based cross-sectional population, with relative ratios (RRs) being 2.40 (95% CI 2.18-2.65) and 2.98 (95% CI 2.62-3.38) compared by LDCT-screening population in U.S. and Europe trials, respectively. MPR of lung cancer is as high as 58.9% in China, with RRs being 0.46 (95% CI 0.31-0.67) and 0.58 (95% CI 0.39-0.85) compared by U.S. and Europe, respectively. CONCLUSIONS: By the epidemiological inference, the LDCT mass screening might be associated with increasing stage I lung cancer and therefore improving population survival outcome. How to translate the experiences of lung cancer screening by LDCT from developed counties to China in a cost-effective manner needs to be further investigated.


Subject(s)
Early Detection of Cancer/methods , Lung Neoplasms/diagnostic imaging , Lung Neoplasms/epidemiology , Tomography, X-Ray Computed/methods , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/diagnostic imaging , Carcinoma, Non-Small-Cell Lung/mortality , China/epidemiology , Europe/epidemiology , Humans , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Mass Screening/methods , Neoplasm Staging , Prognosis , Small Cell Lung Carcinoma/diagnosis , Small Cell Lung Carcinoma/diagnostic imaging , Small Cell Lung Carcinoma/mortality , United States/epidemiology
18.
Oncol Lett ; 11(5): 3501-3507, 2016 May.
Article in English | MEDLINE | ID: mdl-27123142

ABSTRACT

Peritoneal carcinomatosis (PC) of gastric origin is currently recognized as a terminal disease with a poor prognosis. Advancements in novel therapeutic approaches, including intraperitoneal chemotherapy (IPC), have recently been made and it is believed that this may have contributed to the improved survival observed in patients with PC. The present study aimed to investigate overall survival (OS) and the associated prognostic factors in patients with PC of gastric origin who underwent IPC. A total of 57 patients were studied, with a median age of 51 years. The median follow-up time was 12.4 months. PC was diagnosed in all patients with gastric cancer. The median survival time of all patients was 10.1 months, whilst the OS rate at 1, 2 and 3 years was observed to be 46, 19 and 12%, respectively. Symptomatic ascites and a signet ring cell (SRC) histopathological type were demonstrated to signify a poor prognosis. Complete resection of all gross disease (CCR-0) and an increased number of cycles of systemic chemotherapy were independent factors that were observed to correlate with increased OS. The most common morbidities of grade 3/4 adverse effects were bone marrow suppression, nausea or vomiting, and diarrhea. In conclusion, IPC is an important treatment option for patients with PC that has originated from gastric cancer. Symptomatic ascites and SRC adenocarcinoma serve as negative clinicopathological prognostic factors, whilst CCR-0 and increased systemic chemotherapy cycles (≥4 cycles) may prove to be an important therapeutic option for PC patients.

19.
Springerplus ; 5: 368, 2016.
Article in English | MEDLINE | ID: mdl-27066377

ABSTRACT

BACKGROUND: Malignant mesothelioma (MM) is a rare and fatal neoplasm. For diffuse malignant mesothelioma (DMM) patients that were not suitable for cytoreductive surgery and hyperthermic intraperitoneal chemotherapy, systemic chemotherapy is the main treatment. There are no convenient tumor markers to predict the efficacy of treatment and disease progression. This study aimed to evaluate serum CA125 level as a biochemical marker of response to therapy and prognosis in patients with DMM. METHODS: A retrospective study was performed in a single medical institution from April 2008 to April 2014. Overall survival (OS) and prognostic factors were assessed. RESULTS: Forty-one patients were included with a median age of 53 years. The median OS of all patients was 10 months. Patients with baseline CA125 > 280 U/ml had worse OS compared with the patients that baseline CA125 ≤ 280 U/ml. Baseline level of CA125, stage of disease, primary tumor location and systemic chemotherapy were independent prognostic factors associated with OS. In patients who received systemic chemotherapy, the decline in serum CA125 was associated with favorable OS and objective response according to modified Response Evaluation Criteria in Solid Tumors criteria. CONCLUSIONS: The baseline level of serum CA125, accompanied with stage of disease, primary tumor location and systemic chemotherapy, could be regarded as independent prognostic factors for DMM patients. Otherwise, the change in serum CA125 can predict OS and response to systemic chemotherapy.

SELECTION OF CITATIONS
SEARCH DETAIL
...