Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Blood ; 141(7): 766-786, 2023 02 16.
Article in English | MEDLINE | ID: mdl-36322939

ABSTRACT

Extramedullary infiltration (EMI) is a concomitant manifestation that may indicate poor outcome of acute myeloid leukemia (AML). The underlying mechanism remains poorly understood and therapeutic options are limited. Here, we employed single-cell RNA sequencing on bone marrow (BM) and EMI samples from a patient with AML presenting pervasive leukemia cutis. A complement C1Q+ macrophage-like leukemia subset, which was enriched within cutis and existed in BM before EMI manifestations, was identified and further verified in multiple patients with AML. Genomic and transcriptional profiling disclosed mutation and gene expression signatures of patients with EMI that expressed high levels of C1Q. RNA sequencing and quantitative proteomic analysis revealed expression dynamics of C1Q from primary to relapse. Univariate and multivariate analysis demonstrated adverse prognosis significance of C1Q expression. Mechanistically, C1Q expression, which was modulated by transcription factor MAF BZIP transcription factor B, endowed leukemia cells with tissue infiltration ability, which could establish prominent cutaneous or gastrointestinal EMI nodules in patient-derived xenograft and cell line-derived xenograft models. Fibroblasts attracted migration of the C1Q+ leukemia cells through C1Q-globular C1Q receptor recognition and subsequent stimulation of transforming growth factor ß1. This cell-to-cell communication also contributed to survival of C1Q+ leukemia cells under chemotherapy stress. Thus, C1Q served as a marker for AML with adverse prognosis, orchestrating cancer infiltration pathways through communicating with fibroblasts and represents a compelling therapeutic target for EMI.


Subject(s)
Complement C1q , Leukemia, Myeloid, Acute , Humans , Proteomics , Leukemia, Myeloid, Acute/genetics , Leukemia, Myeloid, Acute/metabolism , Bone Marrow/metabolism , Prognosis , Chronic Disease , Recurrence
2.
Gastric Cancer ; 27(2): 387-399, 2024 03.
Article in English | MEDLINE | ID: mdl-38143257

ABSTRACT

BACKGROUND: Data on the long-term oncological outcomes of patients who undergo conversion surgery (CS) in gastric cancer (GC) patients with peritoneal metastasis (PM) are limited. METHODS: GC patients with PM who received intraperitoneal (ip) and systemic chemotherapy between April 2015 and January 2021 were enrolled. Multivariate analysis was performed to identify risk factors associated with survival. Clinicopathological and survival outcomes were compared between those with CS and those without CS (NCS). The paclitaxel (PTX) plus tegafur-gimeracil-oteracil potassium capsules (S-1) (PS) + ip PTX and oxaliplatin plus S-1 (SOX) + ip PTX groups were matched in a 1:1 ratio using propensity score matching. Oncological and survival data were collected and analyzed. RESULTS: A total of 540 patients who received ip chemotherapy via subcutaneous port and systemic chemotherapy were analyzed and 268 patients were enrolled, including 113 who underwent CS and 155 who did not. Overall survival (OS) were 27.0 months and 11.8 months in the CS and NCS groups (P < 0.0001), respectively. R0 resection was an independent prognostic factor for patients who underwent CS. The OS of patients with or without ovariectomy was 21.3 or 12.0 months (P < 0.0001). No difference of clinicopathological and survival outcomes was found between the PS + ip PTX and SOX + ip PTX groups. CONCLUSION: Conversion therapy is safe and adverse events were manageable. CS improves the survival of GC patients with PM after ip and systemic chemotherapy. R0 is an important prognostic factor. Furthermore, outcomes are comparable between the PS + ip PTX and SOX + ip PTX groups.


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Female , Humans , Stomach Neoplasms/pathology , Peritoneal Neoplasms/secondary , Retrospective Studies , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Paclitaxel/therapeutic use
3.
Gastric Cancer ; 26(3): 364-378, 2023 05.
Article in English | MEDLINE | ID: mdl-36738390

ABSTRACT

BACKGROUND: The prognosis of advanced gastric cancer (GC) invading the gastric serosa remains poor, mainly owing to high incidence of peritoneal recurrence. Patients with peritoneal metastases are often treated with neoadjuvant intraperitoneal and systemic chemotherapies (NIPS). Good responders to NIPS often undergo conversion gastrectomy. This study aims to explore biomarkers predicting the occurrence of peritoneal metastasis (PM) and evaluating the efficacy of NIPS in GC patients. METHODS: We collected six peritoneal lavage (PL) samples from two patients with PM, two without PM, and two with diminished PM after NIPS via intraperitoneal access ports. We equally isolated microRNAs from exosomes derived from PL samples for deep sequencing. Two microRNAs (hsa-let-7g-3p and hsa-miR-10395-3p) were identified, and their expression levels were examined in PL samples of 99 GC patients using qRT-PCR. Moreover, we performed in vivo and in vitro functional assays to investigate effects of these microRNAs on metastasis and chemoresistance of GC cells. RESULTS: Exosomal microRNA expression profiling of six PL samples indicated that the microRNA signature in exosomes of PLs from patients with diminished PM was similar to that from patients without PM. Expression levels of hsa-let-7g-3p and hsa-miR-10395-3p were associated with PM. In vivo and in vitro functional assays confirmed that hsa-let-7g-3p and hsa-miR-10395-3p are involved in GC metastasis and chemoresistance. CONCLUSION: PL-derived exosomes in GC contain large amounts of microRNAs related to PM. Moreover, hsa-let-7g-3p and hsa-miR-10395-3p could be used as biomarkers predicting PM and NIPS efficacy and are involved in GC metastasis and chemoresistance.


Subject(s)
Exosomes , MicroRNAs , Peritoneal Neoplasms , Stomach Neoplasms , Humans , Peritoneal Neoplasms/drug therapy , Peritoneal Neoplasms/genetics , Peritoneal Neoplasms/secondary , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology , Peritoneal Lavage , Neoadjuvant Therapy , MicroRNAs/genetics , MicroRNAs/metabolism , Biomarkers , Exosomes/genetics , Exosomes/metabolism , Gene Expression Regulation, Neoplastic
4.
Future Oncol ; 19(38): 2517-2523, 2023 Dec.
Article in English | MEDLINE | ID: mdl-37212686

ABSTRACT

Intraperitoneal chemotherapy combined with systemic chemotherapy is one of the therapeutic modalities currently used for the treatment of gastric cancer patients with peritoneal metastasis. This study was designed to evaluate the efficacy and safety of sintilimab plus S-1 combined intraperitoneal and intravenous paclitaxel. This is an open-label, single-center, phase II study including 36 gastric adenocarcinoma patients with peritoneal metastases diagnosed by laparoscopy. All enrolled patients received sintilimab, intraperitoneal and intravenous paclitaxel plus oral S-1 every 3 weeks. Conversion operation should be considered when a patient responds to the regimen and the peritoneal metastasis disappears. After gastrectomy, the protocol treatment is repeated until disease progression, unacceptable toxicity, investigator decision or patient withdrawal. The primary end point is the 1-year survival rate. Clinical Trial Registration: NCT05204173 (ClinicalTrials.gov).


Subject(s)
Peritoneal Neoplasms , Stomach Neoplasms , Humans , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Neoadjuvant Therapy , Paclitaxel , Peritoneal Neoplasms/secondary , Stomach Neoplasms/pathology
5.
Future Oncol ; 18(10): 1175-1183, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35114800

ABSTRACT

Although recent advances in systemic chemotherapy have improved the clinical outcomes of gastric cancer patients with peritoneal metastasis, the peritoneum still represents a common site of treatment failure and disease recurrence. Neoadjuvant intraperitoneal-systemic chemotherapy has been acknowledged as a more aggressive treatment for gastric cancer patients with peritoneal metastasis. In this multicenter phase III randomized controlled trial, 238 patients will be randomly separated into two groups in a 2:1 ratio after laparoscopic exploration. The experimental arm will receive the proposed neoadjuvant intraperitoneal-systemic chemotherapy regimen, whereas the control group will receive a Paclitaxel + S-1 (PS) chemotherapy regimen. The endpoints for the study are overall survival, response rate, gastrectomy radicality rate, progression-free survival and adverse events.


Recent advances in technology have improved the outcomes of stomach cancer patients. However, there are still many patients who die of cancer that has spread from another part of the body. Neoadjuvant intraperitoneal­systemic chemotherapy has been acknowledged as a more aggressive treatment for stomach cancer patients with peritoneal metastasis (cancer that has spread to the very thin layer of tissue on the inside of the abdomen that covers the stomach and other organs). In this study, 238 patients will be randomly separated into two groups in a 2:1 ratio after evaluation. The experimental group will receive the proposed neoadjuvant intraperitoneal­systemic chemotherapy regimen, whereas the control group will receive a Paclitaxel + S-1 (PS) chemotherapy regimen. The endpoints for the study are how long patients live, number of patients who respond to treatment, number of patients who undergo surgery, how long patients live without their disease getting worse and problems caused by treatment. Trial registration number: ChiCTR-IIR-16009802.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Neoadjuvant Therapy , Peritoneal Neoplasms/secondary , Peritoneal Neoplasms/therapy , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Humans , Neoplasm Staging , Paclitaxel/therapeutic use , Prospective Studies
6.
Future Oncol ; 18(39): 4239-4349, 2022 Dec.
Article in English | MEDLINE | ID: mdl-36651765

ABSTRACT

Although gastric cancer with para-aortic lymph node (PAN) metastasis is commonly regarded as unresectable, surgeons have explored the optimal treatment for patients with PAN metastases limited to No.16a2/b1 in the past few decades. Preoperative systemic therapy combined with D2 gastrectomy plus PAN dissection may improve the prognosis of these patients. In this multicenter phase II trial, 29 gastric cancer patients with PAN metastasis limited to No.16a2/b1 will receive preoperative treatment with nab-paclitaxel, oxaliplatin, S-1 (nab-POS: nab-paclitaxel, oxaliplatin, S-1) and sintilimab followed by D2 gastrectomy plus PAN dissection; and postoperative treatment with oral S-1, intravenous sintilimab and intraperitoneal paclitaxel. The end points for the study are 3-year overall survival, 3-year disease-free survival, pathological response rate, incidence of postoperative complications and adverse events.


Stomach cancer with metastases in the para-aortic lymph nodes is usually considered inoperable. Chemotherapy combined with resection of the stomach and more extensive lymph node dissection may prolong the life of these patients. In this multicenter study, 29 stomach cancer patients with para-aortic lymph node metastases will receive preoperative treatment with nab-paclitaxel, oxaliplatin, S-1 and sintilimab, followed by resection of the stomach combined with para-aortic lymph node dissection and use of continued oral, intravenous and intraperitoneal chemotherapy. The study's end points are 3-year overall survival, 3-year disease-free survival, pathological response rate, incidence of postoperative complications and adverse events. Clinical Trial Registration: ChiCTR2200061125 (ChiCTR.org.cn).


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Lymph Node Excision , Lymphatic Metastasis/pathology , Oxaliplatin , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Lymph Nodes/pathology , Gastrectomy/adverse effects , Multicenter Studies as Topic , Clinical Trials, Phase II as Topic
7.
BMC Cancer ; 21(1): 1344, 2021 Dec 18.
Article in English | MEDLINE | ID: mdl-34922478

ABSTRACT

BACKGROUND: In this study, we tried to access the efficacy and safety of oxaliplatin plus S-1 with intraperitoneal paclitaxel (PTX) for the treatment of Chinese advanced gastric cancer with peritoneal metastases. PATIENTS AND METHODS: Thirty patients diagnosed with advanced gastric cancer underwent laparoscopic exploration and were enrolled when macroscopic disseminated metastases (P1) were confirmed. PTX was diluted in 1 l of normal saline and IP administered through peritoneal port at an initial dose of 40 mg/m2 over 1 h on day1,8, respectively. Oxaliplatin was administered intravenously at an initial dose of 100 mg/m2 on day1, and S-1 was administered orally at an initial dose of 80 mg/m2 for 14 days followed by 7 days rest, repeated by every 3 weeks. RESULTS: Of all these 30 patients, the median number of cycles was 6 (range 2-16) due to the limitation of hematotoxicity and peripheral neuropathy by oxaliplatin. There were 11 (36.7%) patients received conversion surgery. The median progression free survival (PFS) was 6.6 months (95% CI = 4.7-8.5 months) and the median overall survival (OS) was 15.1 months (95% CI = 12.4-17.8 months). The grade 3-4 hematological toxicities were leucopenia (23.3%), neutropenia (23.3%), anemia (16.7%), and thrombocytopenia (20%), respectively. The grade 3-4 non-hematological toxicities were tolerated, most of which were peripheral sensory neuropathy (40%) due to oxaliplatin, diarrhea (20%), nausea and vomiting (26.7%). CONCLUSIONS: SOX+ip PTX regimen was effective in advanced gastric cancer with peritoneal metastasis. Survival time was significantly prolonged by conversion surgery. Grade 3-4 toxicities were uncommon. Large scale clinical trial is necessary to get more evidence to identify its efficacy. TRAIL REGISTRATION: ChiCTR, ChiCTR-IIR-16009802 . Registered 9 November 2016.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Peritoneal Neoplasms/drug therapy , Stomach Neoplasms/drug therapy , Adolescent , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Drug Combinations , Female , Humans , Infusions, Parenteral , Male , Middle Aged , Neoplasm Staging , Oxaliplatin/administration & dosage , Oxaliplatin/adverse effects , Oxonic Acid/administration & dosage , Oxonic Acid/adverse effects , Paclitaxel/administration & dosage , Paclitaxel/adverse effects , Peritoneal Neoplasms/mortality , Peritoneal Neoplasms/secondary , Peritoneum/pathology , Progression-Free Survival , Prospective Studies , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Tegafur/administration & dosage , Tegafur/adverse effects , Young Adult
8.
Future Oncol ; 17(25): 3301-3307, 2021 Sep.
Article in English | MEDLINE | ID: mdl-34008422

ABSTRACT

Although complete omentectomy is traditionally performed in patients with gastric cancer as part of radical gastrectomy to ensure the elimination of micrometastases, the prognostic value of omentectomy during gastrectomy remains unclear. Retrospective studies have shown that the incidence of metastases in the greater omentum is very low in T1-T3 gastric cancer. Thus radical gastrectomy with D2 lymphadenectomy and preservation of the greater omentum may be a proper curative treatment for gastric cancer patients with T1-T3 tumors. The aim of this article is to describe the design and rationale for this prospective, randomized controlled DRAGON-05 trial, conducted to evaluate the prognostic value of omentum-preserving gastrectomy for patients with T1-T3 gastric cancer. Clinical trial registration: ChiCTR2000040045 (ClinicalTrials.gov).


Subject(s)
Gastrectomy/methods , Neoplasm Recurrence, Local/epidemiology , Omentum/surgery , Organ Sparing Treatments/methods , Stomach Neoplasms/surgery , Adolescent , Adult , Aged , Disease-Free Survival , Female , Gastrectomy/statistics & numerical data , Humans , Lymph Node Excision , Male , Middle Aged , Neoplasm Recurrence, Local/prevention & control , Neoplasm Staging , Organ Sparing Treatments/statistics & numerical data , Prognosis , Prospective Studies , Randomized Controlled Trials as Topic , Retrospective Studies , Stomach Neoplasms/diagnosis , Stomach Neoplasms/mortality , Survival Rate , Young Adult
9.
Chin J Cancer Res ; 32(4): 497-507, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32963462

ABSTRACT

OBJECTIVE: Intraperitoneal (IP) chemotherapy through subcutaneous port is an effective treatment for gastric cancer (GC) patients with peritoneal metastasis (PM). The objective of this study is to assess the port complications and risk factors for complications in GC patients with PM. METHODS: In retrospective screening of 301 patients with subcutaneous ports implantation, 249 GC patients with PM who received IP chemotherapy were screened out for analysis. Port complications and risk factors for complications were analyzed. RESULTS: Of the 249 analyzed patients, 57 (22.9%) experienced port complications. Subcutaneous liquid accumulation (42.1%) and infection (28.1%) were the main complications, and other complications included port rotation (14.1%), wound dehiscence (12.3%), inflow obstruction (1.7%) and subcutaneous metastasis (1.7%). The median interval between port implantation and occurrence of complications was 3.0 months. Eastern Cooperative Oncology Group (ECOG) performance status [odds ratio (OR), 1.74; 95% confidence interval (95% CI), 1.12-2.69], albumin (OR, 3.67; 95% CI, 1.96-6.86), implantation procedure optimization (OR, 0.33; 95% CI, 0.18-0.61) and implantation groups (OR, 0.37; 95% CI, 0.20-0.69) were independent risk factors for port complications (P<0.05). ECOG performance status was the only factor that related to the grades of port complications (P=0.016). CONCLUSIONS: Port complications in GC patients who received IP chemotherapy are manageable. ECOG performance status, albumin, implantation procedure and implantation group are independent risk factors for port complications in GC patients with PM.

10.
BMC Cancer ; 14: 10, 2014 Jan 08.
Article in English | MEDLINE | ID: mdl-24401087

ABSTRACT

BACKGROUND: Progesterone is essential for the proliferation and differentiation of mammary gland epithelium. Studies of breast cancer cells have demonstrated a biphasic progesterone response consisting of an initial proliferative burst followed by sustained growth arrest. However, the transcriptional factors acting with the progesterone receptor (PR) to mediate the effects of progesterone on mammary cell growth and differentiation remain to be determined. Recently, it was demonstrated that signal transducer and activator of transcription 6 (Stat6) is a cell growth suppressor. Similar to progesterone-bound PR, Stat6 acts by inducing the expression of the G1 cyclin-dependent kinase inhibitors p21 and p27. The possible interaction between Stat6 and progesterone pathways in mammary cells was therefore investigated in the present study. METHODS: ChIP and luciferase were assayed to determine whether Stat6 induces p21 and p27 expression by recruitment at the proximal Sp1-binding sites of the gene promoters. Immunoprecipitation and Western blotting were performed to investigate the interaction between Stat6 and PR-B. The cellular DNA content and cell cycle distribution in breast cancer cells were analyzed by FACS. RESULTS: We found that Stat6 interacts with progesterone-activated PR in T47D cells. Stat6 synergizes with progesterone-bound PR to transactivate the p21 and p27 gene promoters at the proximal Sp1-binding sites. Moreover, Stat6 overexpression and knockdown, respectively, increased or prevented the induction of p21 and p27 gene expression by progesterone. Stat6 knockdown also abolished the inhibitory effects of progesterone on pRB phosphorylation, G1/S cell cycle progression, and cell proliferation. In addition, knockdown of Stat6 expression prevented the induction of breast cell differentiation markers, previously identified as progesterone target genes. Finally, Stat6 gene expression levels increased following progesterone treatment, indicating a positive auto-regulatory loop between PR and Stat6. CONCLUSIONS: Taken together, these data identify Stat6 as a coactivator of PR mediating the growth-inhibitory and differentiation effects of progesterone on breast cancer cells.


Subject(s)
Breast Neoplasms/genetics , Progesterone/metabolism , Receptors, Progesterone/metabolism , STAT6 Transcription Factor/genetics , Amino Acid Motifs , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Differentiation/genetics , Cell Line, Tumor , Cell Proliferation/genetics , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , Promoter Regions, Genetic , STAT6 Transcription Factor/metabolism
11.
Carcinogenesis ; 34(12): 2851-60, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23803695

ABSTRACT

MPS-1 (metallopanstimulin-1), also known as ribosomal protein S27, was overexpressed in gastric cancer cells. However, how MPS-1 contributes to gastric carcinogenesis has not been well characterized. Here, we show that high expression of MPS-1 was observed in gastric cancer tissues and associated with gastric cancer cell metastasis. Alteration of MPS-1 expression regulates invasion and migration of gastric cancer cells both in vitro and in vivo. Furthermore, by using Signal-Net and cluster analyses of microarray data we identified integrin ß4 (ITGB4) as a downstream target of MPS-1 that mediates its effects on cell metastasis. Knockdown of MPS-1 expression in gastric cancer cells led to significant reduction of ITGB4 expression at both the RNA and protein levels. Mechanically, we found that overexpression of ITGB4 in MPS-1 knockdown cells largely recovers the ability of invasion and migration. Conversely, knockdown of ITGB4 partially reduced cell invading/migrating ability induced by MPS-1 overexpression. Moreover, MPS-1 and ITGB4 expressions are positively correlated in gastric cancer cell lines and tissues. Finally, the survival analyses show that the expression of MPS-1 and ITGB4 is associated with poor outcomes in gastric cancer patients. Collectively, our findings suggest that MPS-1 regulates cell invasiveness and migration partially through ITGB4 and that MPS-1/ITGB4 signaling axis may serve as therapeutic targets in the treatment of gastric cancer.


Subject(s)
Cell Movement/genetics , Integrin beta4/genetics , Integrin beta4/metabolism , Metalloproteins/genetics , Metalloproteins/metabolism , Neoplasm Invasiveness/genetics , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Ribosomal Proteins/genetics , Ribosomal Proteins/metabolism , Stomach Neoplasms/genetics , Adult , Aged , Aged, 80 and over , Cell Line , Cell Line, Tumor , Down-Regulation/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Middle Aged , Neoplasm Invasiveness/pathology , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Stomach Neoplasms/pathology
12.
Redox Biol ; 67: 102923, 2023 11.
Article in English | MEDLINE | ID: mdl-37832398

ABSTRACT

As the predominant immunosuppressive component within the tumor microenvironment (TME), cancer-associated fibroblasts (CAFs) inhibit Natural Killer cell (NK cell) activity to promote tumor progression and immune escape; however, the mechanisms of cross-talk between CAFs and NK cells in gastric cancer (GC) remain poorly understood. In this study, we demonstrate that NK cell levels are inversely correlated with CAFs abundance in human GC. CAFs impair the anti-tumor capacity of NK cells by inducing ferroptosis, a cell death process characterized by the accumulation of iron-dependent lipid peroxides. CAFs induce ferroptosis in NK cells by promoting iron overload; conversely, decreased intracellular iron levels protect NK cells against CAF-induced ferroptosis. Mechanistically, CAFs increase the labile iron pool within NK cells via iron export into the TME, which is mediated by the upregulated expression of iron regulatory genes ferroportin1 and hephaestin in CAFs. Moreover, CAF-derived follistatin like protein 1(FSTL1) upregulates NCOA4 expression in NK cells via the DIP2A-P38 pathway, and NCOA4-mediated ferritinophagy is required for CAF-induced NK cell ferroptosis. In a human patient-derived organoid model, functional targeting of CAFs using a combination of deferoxamine and FSTL1-neutralizing antibody significantly alleviate CAF-induced NK cell ferroptosis and boost the cytotoxicity of NK cells against GC. This study demonstrates a novel mechanism of suppression of NK cell activity by CAFs in the TME and presents a potential therapeutic approach to augment the immune response against GC mediated by NK cells.


Subject(s)
Antineoplastic Agents , Cancer-Associated Fibroblasts , Ferroptosis , Follistatin-Related Proteins , Stomach Neoplasms , Humans , Cancer-Associated Fibroblasts/metabolism , Cancer-Associated Fibroblasts/pathology , Follistatin-Related Proteins/metabolism , Stomach Neoplasms/metabolism , Iron/metabolism , Killer Cells, Natural/metabolism , Killer Cells, Natural/pathology , Antineoplastic Agents/pharmacology , Tumor Microenvironment
13.
Int J Cancer ; 130(12): 2761-70, 2012 Jun 15.
Article in English | MEDLINE | ID: mdl-21796632

ABSTRACT

The ribosomal protein S27 (metallopanstimulin-1, MPS-1) has been reported to be a multifunctional protein, with increased expression in a number of cancers. We reported previously that MPS-1 was highly expressed in human gastric cancer. Knockdown of MPS-1 led to spontaneous apoptosis and repressed proliferation of human gastric cancer cells in vitro and in vivo. However, how does MPS-1 regulate these processes is unclear. Here we performed microarray and pathway analyses to investigate possible pathways involved in MPS-1 knockdown-induced apoptosis in gastric cancer cells. Our results showed that knockdown of MPS-1 inhibited NF-κB activity by reducing phosphorylation of p65 at Ser536 and IκBα at Ser32, inhibiting NF-κB nuclear translocation, and down-regulating its DNA binding activity. Furthermore, data-mining the Gene-Regulatory-Network revealed that growth arrest DNA damage inducible gene 45ß (Gadd45ß), a direct NF-κB target gene, played a critical role in MPS-1 knockdown-induced apoptosis. Over-expression of Gadd45ß inhibited MPS-1 knockdown-induced apoptosis via inhibition of JNK phosphorylation. Taken together, these data revealed a novel pathway, the MPS-1/NF-κB/Gadd45ß signal pathway, played an important role in MPS-1 knockdown-induced apoptosis of gastric cancer cells. This study sheds new light on the role of MPS-1/NF-κB in apoptosis and the possible use of MPS-1 targeting strategy in the treatment of gastric cancer.


Subject(s)
Antigens, Differentiation/metabolism , Apoptosis/genetics , Metalloproteins/metabolism , NF-kappa B/metabolism , Nuclear Proteins/metabolism , RNA-Binding Proteins/metabolism , Ribosomal Proteins/metabolism , Stomach Neoplasms/metabolism , Antigens, Differentiation/biosynthesis , Cell Line, Tumor , Cell Proliferation , Etoposide/pharmacology , HEK293 Cells , Humans , I-kappa B Kinase/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , MAP Kinase Signaling System , Membrane Potential, Mitochondrial , Metalloproteins/genetics , NF-kappa B/biosynthesis , NF-kappa B/genetics , Nuclear Proteins/genetics , Phosphorylation , RNA Interference , RNA, Small Interfering , RNA-Binding Proteins/genetics , Ribosomal Proteins/genetics , Stomach Neoplasms/genetics , Transcription Factor RelA/metabolism
14.
Front Oncol ; 12: 905922, 2022.
Article in English | MEDLINE | ID: mdl-35795055

ABSTRACT

Background: Neoadjuvant intraperitoneal and systemic chemotherapy (NIPS) has shown promising results in gastric cancer (GC) with peritoneal metastasis. However, clinical practice experience of NIPS is still lacking in China. In this study, we investigate the efficacy and safety of NIPS in Chinese patients. Methods: Eligible patients received NIPS every 3 weeks. Gastrectomy was performed for patients who met the criteria of conversion surgery. The primary end point was 1-year overall survival (OS) rate. Secondary end points were the response rate, toxic effects, conversion surgery outcomes and median survival time (MST). Results: Sixty-seven patients were enrolled. The primary endpoint was achieved with 1-year OS rate reached 67.2% (95% CI, 56.8%-79.4%). Conversion surgery was performed in 42 patients (62.9%), and R0 resection was achieved in 23 patients (54.8%) with the MST of 31.3 months (95% CI, 24.3-38.3). And the MST was 19.3 months (95% CI, 16.4-22.2) for all patients. Toxicity and surgical complications were well-tolerated. Moreover, sex, R0 resection, pathological nodal stage and tumor regression grade (TRG) were independent prognostic factors for patients who underwent conversion surgery. Conclusion: The NIPS is effective and safe in treating GC patients with peritoneal metastasis. Male patients, patients who underwent R0 resection, patients with ypN0-1 or TRG 1 after conversion surgery are more likely to benefit from the NIPS. Clinical Trial Registration: http://www.chictr.org.cn/, identifier https://clinicaltrials.gov/ ().

15.
Cancer Sci ; 102(8): 1537-44, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21521417

ABSTRACT

Cyclin D1 is aberrantly expressed in many types of cancers, including breast cancer. High levels of cyclin D1b, the truncated isoform of cyclin D1, have been reported to be associated with a poor prognosis for breast cancer patients. In the present study, we used siRNA to target cyclin D1b overexpression and assessed its ability to suppress breast cancer growth in nude mice. Cyclin D1b siRNA effectively inhibited overexpression of cyclin D1b. Depletion of cyclin D1b promoted apoptosis of cyclin D1b-overexpressing cells and blocked their proliferation and transformation phenotypes. Notably, cyclin D1b overexpression is correlated with triple-negative basal-like breast cancers, which lack specific therapeutic targets. Administration of cyclin D1b siRNA inhibited breast tumor growth in nude mice and cyclin D1b siRNA synergistically enhanced the cell killing effects of doxorubicin in cell culture, with this combination significantly suppressing tumor growth in the mouse model. In conclusion, the results indicate that cyclin D1b, which is overexpressed in breast cancer, may serve as a novel and effective therapeutic target. More importantly, the present study clearly demonstrated a very promising therapeutic potential for cyclin D1b siRNA in the treatment of cyclin D1b-overexpressing breast cancers, including the very malignant triple-negative breast cancers.


Subject(s)
Cyclin D1/antagonists & inhibitors , Mammary Neoplasms, Experimental/therapy , Animals , Apoptosis , Cell Line, Tumor , Cell Proliferation , Doxorubicin/pharmacology , Female , G1 Phase , Humans , Mammary Neoplasms, Experimental/pathology , Mice , Protein Isoforms , RNA, Small Interfering/genetics , Xenograft Model Antitumor Assays
16.
BMC Cancer ; 11: 109, 2011 Mar 27.
Article in English | MEDLINE | ID: mdl-21439087

ABSTRACT

BACKGROUND: Gastric cancer is the second most common cause of global cancer-related mortality. Although dedifferentiation predicts poor prognosis in gastric cancer, the molecular mechanism underlying dedifferentiation, which could provide fundamental insights into tumor development and progression, has yet to be elucidated. Furthermore, the molecular mechanism underlying the effects of hexamethylene bisacetamide (HMBA), a recently discovered differentiation inducer, requires investigation and there are no reported studies concerning the effect of HMBA on gastric cancer. METHODS: Based on the results of FACS analysis, the levels of proteins involved in the cell cycle or apoptosis were determined using western blotting after single treatments and sequential combinations of HMBA and LiCl. GSK-3ß and proton pump were investigated by western blotting after up-regulating Akt expression by Ad-Akt infection. To investigate the effects of HMBA on protein localization and the activities of GSK-3ß, CDK2 and CDK4, kinase assays, immunoprecipitation and western blotting were performed. In addition, northern blotting and RNase protection assays were carried out to determine the functional concentration of HMBA. RESULTS: HMBA increased p27(Kip1) expression and induced cell cycle arrest associated with gastric epithelial cell differentiation. In addition, treating gastric-derived cells with HMBA induced G0/G1 arrest and up-regulation of the proton pump, a marker of gastric cancer differentiation. Moreover, treatment with HMBA increased the expression and activity of GSK-3ß in the nucleus but not the cytosol. HMBA decreased CDK2 activity and induced p27(Kip1) expression, which could be rescued by inhibition of GSK-3ß. Furthermore, HMBA increased p27(Kip1) binding to CDK2, and this was abolished by GSK-3ß inhibition. CONCLUSIONS: The results presented herein suggest that GSK-3ß functions by regulating p27(Kip1) assembly with CDK2, thereby playing a critical role in G0/G1 arrest associated with HMBA-induced gastric epithelial cell differentiation.


Subject(s)
Acetamides/pharmacology , Cyclin-Dependent Kinase Inhibitor p27/metabolism , Glycogen Synthase Kinase 3/metabolism , Intestinal Mucosa/drug effects , Stomach Neoplasms/drug therapy , Aminophenols , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Cycle/drug effects , Cell Differentiation/drug effects , Cell Line, Tumor , Cyclin-Dependent Kinase 2/genetics , Cyclin-Dependent Kinase 2/metabolism , Cyclin-Dependent Kinase Inhibitor p27/genetics , Gene Expression Regulation, Neoplastic , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3 beta , Humans , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Maleimides , Proton Pumps/genetics , Proton Pumps/metabolism , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Stomach Neoplasms/pathology , Stomach Neoplasms/physiopathology
17.
J Cancer ; 12(1): 65-75, 2021.
Article in English | MEDLINE | ID: mdl-33391403

ABSTRACT

Background: Gastric cancer (GC) with peritoneal metastasis has an extremely poor prognosis. Paclitaxel (PTX) intraperitoneal infusion provides an effective treatment for these patients. However, GC patients with peritoneal metastasis who receiving PTX treatments tend to occur PTX-resistance accompany with more aggressive ascites and metastasis. How does this happen is still unknown. Here, we aimed to explore the mechanisms that mediate PTX-resistance and metastasis in GC with peritoneal metastasis. Methods: Ascites samples were collected before PTX infusion and after the relapse in 3 GC patients. To determine the expression of significantly changed proteins, we performed tandem mass tag (TMT) quantitative proteomics. Immunohistochemistry (IHC) staining and western blot were performed to confirm the expression of CDH11 in the PTX-resistant tissues and MKN45P-PR cells. Invasion and migration of GC cells were examined by in vitro transwell and wound healing assays and in vivo dissemination experiments. Results: CDH11 expression was downregulated in the relapsed PTX-resistant ascites, tissues and the PTX-resistant cell line MKN45P-PR. Inhibition of CDH11 expression promoted the invasion, migration and PTX resistance of MKN45P cells, while overexpression of CDH11 repressed these biological functions. Moreover, tumors disseminated in the mice peritoneal cavity induced by MKN45P-PR cells and shCDH11 cells displayed higher metastatic ability and resistance to PTX treatment. Conclusions: Our results reveal that CDH11 is inhibited in the relapsed PTX-resistant patients and the downregulated CDH11 expression promotes GC cell invasion, migration and PTX resistance. CDH11 may have the potential to serve as a predictable marker for the occurrence of PTX resistance in GC patients with peritoneal metastasis.

18.
Cancer Biomark ; 31(4): 329-338, 2021.
Article in English | MEDLINE | ID: mdl-33896831

ABSTRACT

BACKGROUND: Gastric cancer (GC) patients with peritoneal metastasis usually have extremely poor prognosis. Intraperitoneal infusion of paclitaxel (PTX) provides an effective treatment, but relapse and PTX-resistance are unavoidable disadvantages, and it is difficult to monitor the occurrence of PTX-resistance. OBJECTIVE: The aim of this study was to explore novel autoantibodies in the ascites of individuals with relapsed PTX-resistant GC with peritoneal metastasis. METHODS: Ascites samples were collected before PTX infusion and after the relapse in 3 GC patients. To determine the expression of significantly changed proteins, we performed autoantibody profiling with immunome protein microarrays and tandem mass tag (TMT) quantitative proteomics, and then, the overlapping proteins were selected. RESULTS: Thirty-eight autoantibodies that were differentially expressed between the ascites in the untreated group and relapsed PTX-resistant group were identified. For confirmation of the results, TMT quantitative proteomics was performed, and 842 dysregulated proteins were identified. Four proteins, TPM3, EFHD2, KRT19 and vimentin, overlapped between these two assays. CONCLUSIONS: Our results first revealed that TPM3, EFHD2, KRT19 and vimentin were novel autoantibodies in the ascites of relapsed PTX-resistant GC patients. These autoantibodies may be used as potential biomarkers to monitor the occurrence of PTX-resistance.


Subject(s)
Ascites/immunology , Autoantibodies/analysis , Peritoneal Neoplasms/secondary , Stomach Neoplasms/immunology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Autoantibodies/immunology , Drug Resistance, Neoplasm , Humans , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Peritoneal Neoplasms/immunology , Peritoneal Neoplasms/pathology , Protein Array Analysis/methods , Proteomics/methods , Stomach Neoplasms/drug therapy , Stomach Neoplasms/pathology
19.
Am J Transl Res ; 13(3): 1568-1579, 2021.
Article in English | MEDLINE | ID: mdl-33841680

ABSTRACT

BACKGROUND: Neoadjuvant chemotherapy (NAC) plays an important role in the therapeutic strategy of locally advanced gastric cancer (LAGC). However, the response of LAGC after NAC varies among different patients. The objective response after NAC has proven to be an excellent indicator for benefiting from NAC, yet effective predictors of objective response are still lacking. The present study aimed to identify potential predictors of objective response in LAGC patients treated with NAC. METHODS: Clinicopathological data from 267 patients with LAGC who received NAC and met the inclusion criteria between July 2009 and December 2018 were retrospectively reviewed. Patients were randomly divided into the training and test sets at a 2:1 ratio. Univariate analysis was used to investigate whether any factors were correlated with objective response in the training set. Multivariate logistic regression analysis was applied to find independent predictors. A risk score model was then constructed based on the independent predictors, and its performance in predicting objective response was validated in the test set. RESULTS: Univariate analysis found that gender, age, short axis diameter of the largest regional lymph node (LNmax), serum total protein content, CEA detection value, tumor location, tumor differentiation, signet ring cell carcinoma component and Borrmann type were potential predictors for objective response. In multivariate logistic regression analysis, gender, LNmax and signet ring cell carcinoma component were independent predictors for objective response. Based on independent predictors, we developed a prediction model for objective response. CONCLUSIONS: We found gender, LNmax and signet ring cell carcinoma component were independent predictors for objective response. The prediction model is a good tool to predict the objective response for LAGC patients treated with NAC, which can be applied to guide clinical practice.

20.
Redox Biol ; 46: 102076, 2021 10.
Article in English | MEDLINE | ID: mdl-34315112

ABSTRACT

Acquired resistance to tyrosine kinase inhibitors (TKIs) is the major obstacle to improve clinical efficacy in cancer patients. The epithelial-stromal interaction in tumor microenvironment influences cancer drug response to TKIs. Anlotinib is a novel oral multi-targeted TKI, and has recently been proven to be effective and safe for several tumors. However, if and how the epithelial-stromal interaction in tumor microenvironment affects anlotinib response in gastric cancer (GC) is not known. In this study, we found that anlotinib inhibited GC cells growth by inducing GC cells apoptosis and G2/M phase arrest in a dose- and time-dependent manner. Reactive oxygen species (ROS) mediated anlotinib-induced apoptosis in GC cells, while cancer-associated fibroblasts (CAFs) significantly suppressed anlotinib-induced apoptosis and ROS in GC cells. Increased BDNF that was derived from CAFs activated TrkB-Nrf2 signaling in GC cells, and reduced GC cells response to anlotinib. We identified secreted lactate from GC cells as the key molecule instructing CAFs to produce BDNF in a NF-κB-dependent manner. Additionally, functional targeting BDNF-TrkB pathway with neutralizing antibodies against BDNF and TrkB increased the sensitivity of GC cells towards anlotinib in human patient-derived organoid (PDO) model. Taken together, these results characterize a critical role of the epithelial-stroma interaction mediated by the lactate/BDNF/TrkB signaling in GC anlotinib resistance, and provide a novel option to overcome drug resistance.


Subject(s)
Brain-Derived Neurotrophic Factor , Stomach Neoplasms , Brain-Derived Neurotrophic Factor/genetics , Fibroblasts , Humans , Indoles , Lactic Acid , Quinolines , Stomach Neoplasms/drug therapy , Stomach Neoplasms/genetics , Tumor Microenvironment
SELECTION OF CITATIONS
SEARCH DETAIL