Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 63
Filter
1.
Clin Transl Med ; 14(2): e1578, 2024 02.
Article in English | MEDLINE | ID: mdl-38356419

ABSTRACT

BACKGROUND AND AIMS: In gastric cancer, the response rate of programmed cell death protein-1 (PD-1) inhibitor is far from satisfactory, indicating additional nonredundant pathways might hamper antitumour immunity. V-domain immunoglobulin suppressor of T-cell activation (VISTA) has been reported in several malignancies as a novel immune-checkpoint. Nevertheless, the role of VISTA in gastric cancer still remains obscure. Our purpose is to explore the clinical significance and potential mechanism of VISTA in affecting gastric cancer patients' survival and immunotherapeutic responsiveness. METHODS: Our study recruited eight independent cohorts with a total of 1403 gastric cancer patients. Immunohistochemistry, multiplex immunofluorescence, flow cytometry or intracellular flow cytometry, quantitative polymerase chain reaction, western blotting, fluorescence-activated cell sorting, magnetic-activated cell sorting, smart-seq2, in vitro cell co-culture and ex vivo tumour inhibition assays were applied to investigate the clinical significance and potential mechanism of VISTA in gastric cancer. RESULTS: VISTA was predominantly expressed on tumour-associated macrophages (TAMs), and indicated poor clinical outcomes and inferior immunotherapeutic responsiveness. VISTA+ TAMs showed a mixed phenotype. Co-culture of TAMs and CD8+ T cells indicated that VISTA+ TAMs attenuated effective function of CD8+ T cells. Blockade of VISTA reprogrammed TAMs to a proinflammatory phenotype, reactivated CD8+ T cells and promoted apoptosis of tumour cells. Moreover, blockade of VISTA could also enhance the efficacy of PD-1 inhibitor, suggesting that blockade of VISTA might synergise with PD-1 inhibitor in gastric cancer. CONCLUSIONS: Our data revealed that VISTA was an immune-checkpoint associated with immunotherapeutic resistance. Blockade of VISTA reprogrammed TAMs, promoted T-cell-mediated antitumour immunity, and enhanced efficacy of PD-1 inhibitor, which might have implications in the treatment of gastric cancer.


Subject(s)
Stomach Neoplasms , Humans , Stomach Neoplasms/pathology , CD8-Positive T-Lymphocytes , B7-H1 Antigen/metabolism , Immune Checkpoint Inhibitors , Tumor-Associated Macrophages/metabolism , Immunoglobulins
2.
BMC Cancer ; 23(1): 308, 2023 Apr 04.
Article in English | MEDLINE | ID: mdl-37016303

ABSTRACT

BACKGROUND: The Neo-REGATTA study evaluated the effectiveness and safety of Docetaxel, oxaliplatin, and S-1 (DOS regimen) followed by radical resection vs. chemotherapy in advanced gastric adenocarcinoma patients with single non-curable factor. METHODS: This cohort study prospectively enrolled advanced gastric adenocarcinoma patients with single non-curable factor between November 2017 and June 2021. Patients without progression after four cycles of DOS were divided into resection group and chemotherapy group. The outcomes included overall survival (OS), progression-free survival (PFS) and safety. Effectiveness analysis was also performed by propensity score matching (PSM). RESULTS: A total of 73 patients were enrolled and 13 patients were withdrawn due to disease progression after 4 cycles of DOS. Afterwards, 35 and 25 participants were in the resection and chemotherapy groups, respectively. After a median follow-up time of 30.0 months, the median PFS and OS were 9.0 months, and 18.0 months for the chemotherapy group, but not reached in the resection group. After PSM, 19 matched participants were in each group, and the median PFS and OS were longer in resection group than that in chemotherapy group. The most common grade 3 or 4 adverse events both in the resection group and chemotherapy groups were neutropenia (5.7%, 8.0%) and leukopenia (5.7%, 8.0%). CONCLUSIONS: Radical resection might provide survival benefit compared with continuous chemotherapy alone in advanced gastric adenocarcinoma patients who had a disease control after DOS, with a good safety profile. TRIAL REGISTRATION: The study protocol was registered on ClinicalTrial.gov (NCT03001726, 23/12/2016).


Subject(s)
Adenocarcinoma , Stomach Neoplasms , Humans , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Stomach Neoplasms/pathology , Docetaxel/therapeutic use , Oxaliplatin/therapeutic use , Neoadjuvant Therapy , Cohort Studies , Prospective Studies , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Adenocarcinoma/pathology
3.
Nat Commun ; 13(1): 6807, 2022 11 10.
Article in English | MEDLINE | ID: mdl-36357415

ABSTRACT

The synergistic effect of neoadjuvant immunotherapy and chemoradiotherapy in gastric adenocarcinoma is unclear. This phase II trial (NCT03631615) investigated this neoadjuvant combination in locally advanced adenocarcinoma of stomach or gastroesophageal junction. Thirty-six patients received capecitabine 850 mg/m2 twice daily and simultaneous radiotherapy for 5 weeks, sandwiched by a 21-day cycle of oxaliplatin 130 mg/m2 (day 1) plus capecitabine 1000 mg/m2 twice daily (days 1-14), respectively, followed by surgery. Camrelizumab 200 mg (day 1) was given for 5 cycles since initiating chemotherapy. Primary endpoint was pathological complete response (pCR, ypT0) rate. Secondary endpoints included total pCR (tpCR, ypT0N0) rate, major pathological response (MPR, < 10% residual tumor cells) rate, margin-free (R0) resection rate, downstaging, progression-free survival (PFS), overall survival (OS), and safety. The pCR rate was 33.3% (95% CI, 18.6-51.0), meeting pre-specified endpoint. TpCR, MPR, and R0 resection rates were 33.3%, 44.4%, and 91.7%, respectively. Twenty-eight (77.8%) patients reached ypN0. Two-year PFS and OS rates were 66.9% and 76.1%, respectively. The most common grade 3-4 adverse event was decreased lymphocyte count (27 [75.0%]). Neoadjuvant camrelizumab plus concurrent chemoradiotherapy exhibits promising pathological response in patients with locally advanced gastric adenocarcinoma, with an acceptable safety profile.


Subject(s)
Adenocarcinoma , Rectal Neoplasms , Stomach Neoplasms , Humans , Neoadjuvant Therapy/adverse effects , Capecitabine/therapeutic use , Planets , Rectal Neoplasms/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Chemoradiotherapy/adverse effects , Adenocarcinoma/drug therapy , Esophagogastric Junction/pathology , Stomach Neoplasms/drug therapy
4.
Cell Death Discov ; 8(1): 321, 2022 Jul 14.
Article in English | MEDLINE | ID: mdl-35835735

ABSTRACT

Human epidermal growth factor receptor 2 (HER-2), a famous therapeutic target for breast cancer, is also associated with an increased risk of recurrence and poor outcomes of other malignancies, including gastric cancer. Yet the mechanism of HER-2 therapy resistance remains controversial due to the heterogeneity of gastric adenocarcinoma. We know, Procollagen-Lysine,2-Oxoglutarate 5-Dioxygenase 3 (PLOD3), a key gene coding enzymes that catalyze the lysyl hydroxylation of extracellular matrix collagen, plays an important contributor to HER-2 targeting agent Trastuzumab resistance in gastric cancer. Herein, we analyzed clinical samples of gastric cancer patients and gastric cancer cell lines and identified PLOD3, unveiled that depletion of PLOD3 leads to decreased cell proliferation, tumor growth and Trastuzumab sensitivity in these Trastuzumab resistant GC cell lines. Clinically, increased PLOD3 expression correlates with decreased Trastuzumab therapy responsiveness in GC patients. Mechanistically, we show that PLOD3 represses tumor suppressor FoxO3 expression, therefore upregulating Survivin protein expression that contributes to Trastuzumab resistance in GC. Therefore, our study identifies a new signaling axis PLOD3-FoxO3- Survivin pathway that may be therapeutically targeted in HER-2 positive gastric cancer.

5.
BMC Med ; 20(1): 107, 2022 04 06.
Article in English | MEDLINE | ID: mdl-35382819

ABSTRACT

BACKGROUND: Adding anti-angiogenics to neoadjuvant chemotherapy for localized gastric cancer is recognized as a promising strategy, but its clinical value remains to be defined. METHODS: This single-center, single-arm, phase 2 trial included patients with locally advanced (cT3/4aN+M0) adenocarcinoma of the stomach or gastroesophageal junction (GEJ) who received three cycles of intravenous oxaliplatin (135 mg/m2 on day 1), oral capecitabine (1000 mg/m2 twice daily on days 1 to 14), and oral apatinib for 21 days (250 mg once daily in the first two cycles, and further increased to 500 mg daily in the third cycle based on whether any adverse event of grade 3 or worse occurred), and an additional cycle of oxaliplatin plus capecitabine, followed by gastrectomy with D2 lymphadenectomy. The primary endpoint was the proportion of patients who achieved an objective response according to RECIST version 1.1. RESULTS: Between April 28, 2017, and October 23, 2019, 37 patients were screened and 35 participants were included. Of the 32 patients assessable for efficacy and safety, objective responses were achieved in 25 (78.1%; 95% confidence interval [CI], 60.0% to 90.7%) patients. Thirty-one (96.9%) patients received R0 resection, two (6.3%) patients achieved pathological complete response, and 11 (34.4%) patients achieved pathological response. At the data cutoff date (September 30, 2021), the median event-free survival was 42.6 (95% CI, 16.2 to not reached) months, and the median overall survival was not reached. The most common grade 3 or 4 treatment-emergent adverse events were hypertension (9/32, 28.1%), thrombocytopenia (7/32, 21.9%), and neutropenia (5/32, 15.6%). Seven (21.9%) patients developed surgical complications, and the most common one was intra-abdominal abscess (4/32, 12.5%). CONCLUSIONS: The concomitant use of apatinib, oxaliplatin, and capecitabine as neoadjuvant therapy showed promising efficacy and manageable safety profile in patients with locally advanced adenocarcinoma of the stomach or GEJ, and further phase 3 study is warranted. TRIAL REGISTRATION: This study was registered with ClinicalTrial.gov ( NCT03229096 ).


Subject(s)
Adenocarcinoma , Neoadjuvant Therapy , Adenocarcinoma/pathology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine/adverse effects , Esophagogastric Junction/pathology , Humans , Oxaliplatin/adverse effects , Pyridines
6.
J Exp Clin Cancer Res ; 41(1): 14, 2022 Jan 07.
Article in English | MEDLINE | ID: mdl-34996504

ABSTRACT

BACKGROUND: Chemoresistance is a main obstacle in gastric cancer (GC) treatment, but its molecular mechanism still needs to be elucidated. Here, we aim to reveal the underlying mechanisms of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) resistance in GC. METHODS: We performed RNA sequencing (RNA-seq) on samples from patients who were resistant or sensitive to nab-paclitaxel, and identified Zinc Finger Protein 64 (ZFP64) as critical for nab-paclitaxel resistance in GC. CCK8, flow cytometry, TUNEL staining, sphere formation assays were performed to investigate the effects of ZFP64 in vitro, while subcutaneous tumor formation models were established in nude mice or humanized mice to evaluate the biological roles of ZFP64 in vivo. Chromatin immunoprecipitation sequencing (CHIP-seq) and double-luciferase reporter gene assay were conducted to reveal the underlying mechanism of ZFP64. RESULTS: ZFP64 overexpression was linked with aggressive phenotypes, nab-paclitaxel resistance and served as an independent prognostic factor in GC. As a transcription factor, ZFP64 directly binds to Galectin-1 (GAL-1) promoter and promoted GAL-1 transcription, thus inducing stem-cell like phenotypes and immunosuppressive microenvironment in GC. Importantly, compared to treatment with nab-paclitaxel alone, nab-paclitaxel plus GAL-1 blockade significantly enhanced the anti-tumor effect in mouse models, particularly in humanized mice. CONCLUSIONS: Our data support a pivotal role for ZFP64 in GC progression by simultaneously promoting cellular chemotherapy resistance and tumor immunosuppression. Treatment with the combination of nab-paclitaxel and a GAL-1 inhibitor might benefit a subgroup of GC patients.


Subject(s)
Albumins/therapeutic use , DNA-Binding Proteins/metabolism , Nuclear Proteins/metabolism , Paclitaxel/therapeutic use , Stomach Neoplasms/drug therapy , Transcription Factors/metabolism , Albumins/pharmacology , Animals , Cell Movement , Humans , Male , Mice , Paclitaxel/pharmacology , Prospective Studies , Stomach Neoplasms/pathology , Transfection , Tumor Microenvironment , Xenograft Model Antitumor Assays
7.
Ann Surg ; 275(1): e163-e173, 2022 01 01.
Article in English | MEDLINE | ID: mdl-32511132

ABSTRACT

OBJECTIVE: To examine the clinical significance of LAP to predict survival outcomes and chemotherapeutic responsiveness in gastric cancer. BACKGROUND: LAP has been shown to possess significant immunoregulatory roles in several malignancies. However, the role and clinical significance of LAP in gastric cancer still remains unknown. METHODS: Four hundred and fifty-six tumor tissue microarray specimens, 80 fresh tumor tissue samples of gastric cancer patients from Zhongshan Hospital, Fudan University and transcriptomic and clinical data of 328 gastric cancer patients from the Cancer Genome Atlas were analyzed. LAP expression and immune contexture were examined by immunohistochemistry, CIBERSORT, and flow cytometry. Clinical outcomes of patient subgroups were compared by Kaplan-Meier curves, Cox model and interaction test. RESULTS: High LAP expression predicted poor overall survival (P < 0.001, P < 0.001, and P = 0.022) and inferior therapeutic responsiveness to fluorouracil-based adjuvant chemotherapy (P = 0.008 for interaction) in gastric cancer. LAP was associated with immunoevasive tumor microenvironment featured by dysfunctional CD8+ T cells infiltration (P < 0.001). The LAP-associated dysfunctional CD8+ T cells had an exhausted phenotype with decreased effector molecules such as interferon-γ, granzyme B, and perforin, but also elevated programmed cell death protein-1, which resulted in poor prognosis and inferior therapeutic responsiveness. CONCLUSIONS: This study revealed that LAP could identify immunoevasive subtype gastric cancer, indicating LAP might be a potential immunotherapeutic target and facilitate patient counseling on individualized adjuvant therapy and follow-up scheduling in gastric cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Neoplasm Staging , Peptides/metabolism , Stomach Neoplasms/metabolism , Tumor Microenvironment/immunology , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant/methods , Female , Gastrectomy , Humans , Male , Middle Aged , Prognosis , Stomach Neoplasms/diagnosis , Stomach Neoplasms/therapy
8.
Endocr Connect ; 10(8): 947-954, 2021 Aug 13.
Article in English | MEDLINE | ID: mdl-34289448

ABSTRACT

A new subcategory, grade 3 neuroendocrine tumors, is incorporated into the grading system of pancreatic neuroendocrine neoplasms in the 2017 WHO classification in order to differentiate grade 3 neuroendocrine tumors from neuroendocrine carcinomas. The 2019 WHO classification extends the concept of grade 3 neuroendocrine tumors to gastrointestinal high-grade neuroendocrine neoplasms. However, there is still limited study focusing on the gastric grade 3 neuroendocrine tumors and gastric neuroendocrine carcinomas. We retrospectively enrolled 151 gastric high-grade neuroendocrine neoplasms patients, who underwent radical resection from January 2007 to December 2015. Clinicopathologic and prognostic features were studied. The Surveillance, Epidemiology, and End Results (SEER) database was used to verify the prognostic determinants found in the Zhongshan cohort. Neuroendocrine carcinomas showed a higher Ki67 index and higher mitotic count than grade 3 neuroendocrine tumors. We identified 109 (72.2%) patients with neuroendocrine carcinomas, 12 (7.9%) patients with grade 3 neuroendocrine tumors, and 30 (19.9%) patients with mixed neuroendocrine-non-neuroendocrine neoplasms. Although neuroendocrine carcinomas demonstrated higher Ki67 index (P = 0.004) and mitoses (P = 0.001) than grade 3 neuroendocrine tumors, their prognosis after radical resection did not demonstrate significant differences (P = 0.709). Tumor size, perineural invasion, and TNM stage were independent prognostic factors of gastric high-grade neuroendocrine neoplasms.

9.
Oncoimmunology ; 9(1): 1856468, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33354409

ABSTRACT

Interleukin-9 (IL-9) is a T cell cytokine that is associated with inflammation and allergy, but the expression level of IL-9 in gastric cancer and its clinical significance are less well established. Our study aims to uncover the critical role of IL-9 in the progression of gastric cancer. Here, a total of 453 patients with gastric cancer undergoing curative resection were enrolled for immunohistochemical analyses, and Kaplan-Meier analysis was conducted to compare overall survival of patients in different subgroups. We further investigated the correlation between IL-9 expression and functional status of intratumoral CD8+ T cells by means of Flow cytometry. Moreover, in vitro study was preformed to further explore the influence of IL-9 on anti-tumor immunity. Results indicated that gastric cancer patients with high IL-9 expression showed improved overall survival and gained more benefit from 5-fluorouracil-based adjuvant chemotherapy (ACT). High IL-9 expression was associated with increased numbers and elevated function of intratumoral CD8+ T cells. In vitro study revealed that recombinant human IL-9 (rhIL-9) exhibit anti-tumor activity via enhancing the function of intratumoral CD8+ T cells. Moreover, we found rhIL-9 could augment the efficacy of Pembrolizumab in gastric cancer. In summary, these results suggest that IL-9 expression could act as an independent predictor for overall survival and ACT response and enhancing IL-9 signaling might represent an important therapeutic strategy in gastric cancer.


Subject(s)
Interleukin-9 , Stomach Neoplasms , CD8-Positive T-Lymphocytes , Chemotherapy, Adjuvant , Humans , Prognosis , Stomach Neoplasms/drug therapy
10.
J Surg Res ; 255: 285-296, 2020 11.
Article in English | MEDLINE | ID: mdl-32574755

ABSTRACT

BACKGROUND: The necessity of extensive lymph node (LN) dissection/examination and adjuvant therapy for patients with early gastric cancer (EGC, Tis-T1, any N) remains controversial. We aim to refine treatment recommendations for patients with EGC through a reflective analysis for the survival gap between Eastern and Western countries. METHODS: EGC patients diagnosed between 2004 and 2014 were identified from the National Cancer Database (NCDB) and a large medical center in China. Adequate LN yield was defined as ≥25 LNs examined. RESULTS: In the US cohort, 14.4% of (1104/7641) patients with EGC had ≥25 LNs examined. The 5-y overall survival (OS) was significantly better than those with <25 LNs (78.9% versus 68.5%, P < 0.001). Examination of ≥25 LNs was an independent predictor of better OS after adjusting all known prognostic factors. Patients with ≥25 LNs examined had significantly higher chance of having LN-positive disease compared to patients with <25 LNs (14.9% versus 10.7%, P < 0.001). A similar stage migration phenomenon was observed in Chinese cohort (LN positive: 25.2% versus 18.4% in ≥25 LNs and <25 LNs examined group, respectively, P = 0.02). In the US cohort, adjuvant therapy was associated with a significant survival benefit for LN-positive patients (5-y OS: 71.0% versus 43.0% for with/without adjuvant therapy, respectively, P < 0.001) but not in LN-negative patients (5-y OS: 71.2% versus 71.5%, P = 0.90). CONCLUSIONS: Adequate lymphadenectomy and LN examination are critical components of EGC management. Adjuvant therapy should be strongly encouraged for all EGC patients with LN-positive disease in the United States.


Subject(s)
Carcinoma/surgery , Lymph Node Excision/statistics & numerical data , Stomach Neoplasms/surgery , Aged , Aged, 80 and over , Carcinoma/mortality , China/epidemiology , Combined Modality Therapy , Female , Humans , Male , Middle Aged , Retrospective Studies , Stomach Neoplasms/mortality , United States/epidemiology
11.
Br J Cancer ; 122(10): 1525-1534, 2020 05.
Article in English | MEDLINE | ID: mdl-32205862

ABSTRACT

BACKGROUND: Intratumoural CD103+CD8+ T cells have been linked to prolonged survival in several malignancies. However, the clinical significance of CD103+CD8+ T cells in gastric cancer remains unexplored. METHODS: Gastric cancer tissues from Zhongshan Hospital and data from Gene Expression Omnibus were obtained and analysed. Immunohistochemistry and flow cytometry were performed to detect the number and phenotypical characteristics of CD103+CD8+ T cells. The effect of programmed cell death protein-1 (PD-1) blockade on CD103+CD8+ T cells was evaluated with the use of an in vitro study based on fresh tumour tissues. RESULTS: CD103+CD8+ T cells predicted superior overall survival and provided better prognostic power than total CD8+ T cells in gastric cancer. Patients with high CD103+CD8+ T cell infiltration also gained more benefit from adjuvant chemotherapy. Flow cytometry analysis showed that CD103+CD8+ T cells exerted superior anti-tumour effects with stronger retention capacity and cytotoxicity. Moreover, an in vitro study showed that CD103+CD8+ T cells were more functionally restored after PD-1 blockade than CD103-CD8+ T cells. CONCLUSIONS: CD103+CD8+ T cells might be a useful marker to predict prognosis and therapeutic efficacy for gastric cancer patients. Efforts to increase intratumoural CD103+CD8+ T cell frequency might be a novel therapeutic strategy in gastric cancer.


Subject(s)
Antigens, CD/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Lineage/immunology , Integrin alpha Chains/immunology , Stomach Neoplasms/immunology , Aged , Female , Gene Expression Regulation, Neoplastic/genetics , Gene Expression Regulation, Neoplastic/immunology , Humans , Kaplan-Meier Estimate , Lymphocytes, Tumor-Infiltrating , Male , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/genetics , Stomach Neoplasms/classification , Stomach Neoplasms/genetics , Stomach Neoplasms/pathology
12.
Cancer Immunol Immunother ; 69(7): 1327-1336, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32200421

ABSTRACT

With dichotomous etiology and pathogenesis, intestinal type and diffuse type gastric cancers vary in their clinical and molecular features to the point of representing distinct entities. However, the differences of tumor-infiltrating immune cells within the two types of gastric cancer have not been well researched. This study was aimed to evaluate the functional impact of Lauren classification on immune contexture in gastric cancer patients. Tumor tissues of gastric cancer patients from Zhongshan Hospital and gastric cancer data from The Cancer Genome Atlas (TCGA) cohort were analyzed. By immunohistochemistry and flow cytometry, we found that intratumoral CD8+ T cells were more abundant but less functional in diffuse type as compared with those in intestinal type tumor tissues. Survival analysis indicated that CD8+ T cells yielded favorable prognosis only in intestinal type patients other than diffuse type cancer patients. Moreover, such diffuse type-associated CD8+ T cell dysfunction was featured by elevated expression of immunosuppressive factors including interleukin-10 (IL-10), transforming growth factor-ß1 (TGF-ß1) and indoleamine 2,3-dioxygenase 1 (IDO1). In summary, we found that the density, prognostic significance and functional status of intratumoral CD8+ T cells varied with Lauren subtypes in gastric cancer. These results further indicated Lauren classification might be a potential therapeutic marker, and should be considered in therapeutic decisions, especially immunotherapeutic eligibility.


Subject(s)
Adenocarcinoma/classification , Adenocarcinoma/immunology , Biomarkers, Tumor/analysis , CD8-Positive T-Lymphocytes/immunology , Stomach Neoplasms/classification , Stomach Neoplasms/immunology , Adenocarcinoma/pathology , Follow-Up Studies , Humans , Prognosis , Stomach Neoplasms/pathology
13.
Eur J Cancer ; 128: 27-37, 2020 03.
Article in English | MEDLINE | ID: mdl-32109848

ABSTRACT

AIM: Tumour-associated macrophages (TAMs) are prominent immune cells infiltrating in solid tumours with phenotypic and functional heterogeneity. However, the clinical significance of heterogeneous subtypes of TAMs in gastric cancer still remains obscure. Here, we aimed to explore the clinical significance of TAMs expressing dendritic cell-specific intercellular adhesion molecule 3-grabbing non-integrin (DC-SIGN) and its relevance with immune contexture in gastric cancer. METHODS: We selected 453 formalin-fixed and paraffin-embedded samples and 51 fresh tissue specimens of patients with gastric cancer from Zhongshan Hospital. The association of DC-SIGN+ macrophages with clinicopathological parameters, overall survival (OS) and responsiveness to fluorouracil-based adjuvant chemotherapy (ACT) was inspected. Immunohistochemistry (IHC) and flow cytometry (FCM) were applied to characterize immune cells in gastric cancer. RESULTS: We demonstrated that high intratumoral DC-SIGN+ macrophages infiltration predicted poor OS and inferior therapeutic responsiveness to fluorouracil-based ACT in patients with gastric cancer. Furthermore, higher infiltration of DC-SIGN+ macrophages indicated an increased number of Foxp3+ regulatory T cells (Tregs), CD8+ T cells and a higher ratio of Foxp3+/CD8+ within the tumour microenvironment (TME). In addition, CD8+ T cells in DC-SIGN+ macrophages high subgroup were functionally impaired, showing decreased interferon-γ (IFN-γ), granzyme B (GZMB) and perforin production yet elevated programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) expression. CONCLUSIONS: DC-SIGN+ macrophages were associated with immunoinvasive TME and indicated poor prognosis and inferior therapeutic responsiveness to fluorouracil-based ACT. DC-SIGN+ macrophages might be an independent prognosticator and a potential immunotherapeutic target for gastric cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cell Adhesion Molecules/metabolism , Lectins, C-Type/metabolism , Macrophages/immunology , Receptors, Cell Surface/metabolism , Stomach Neoplasms/immunology , Tumor Escape , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Chemotherapy, Adjuvant/methods , Drug Resistance, Neoplasm/immunology , Female , Fluorouracil/pharmacology , Fluorouracil/therapeutic use , Follow-Up Studies , Gastrectomy , Humans , Kaplan-Meier Estimate , Macrophages/metabolism , Male , Middle Aged , Stomach/cytology , Stomach/immunology , Stomach/pathology , Stomach/surgery , Stomach Neoplasms/mortality , Stomach Neoplasms/pathology , Stomach Neoplasms/therapy , Treatment Outcome
14.
Front Oncol ; 10: 574937, 2020.
Article in English | MEDLINE | ID: mdl-33479597

ABSTRACT

INTRODUCTION: Although traditional treatments confer survival benefits to patients with gastric cancer (GC), many patients experience relapse soon after postoperative adjuvant therapy. Immune-related mechanisms play an important role in GC, and immunotherapeutic strategies are considered to be a promising direction for the treatment of GC. Thus, our study aimed to investigate the expression and prognostic significance of immune-related genes in GC. METHODS: Formalin-fixed, paraffin-embedded samples were collected from 48 resectable GC patients. The transcriptome data of the tumor immune microenvironment were assessed using an immuno-oncology 395-gene panel RNA sequencing platform. The prognostic value of the 395 genes was analyzed and validated in the KM plotter and GEPIA databases. The data from The Cancer Genome Atlas (TCGA, downloaded from UCSC Xena repository) and Tumor IMmune Estimation Resource (TIMER) were used to evaluate the correlations between prognostic factors and immune signatures. RESULTS: Among the 395 genes, NOTCH3 was identified as a good prognostic factor for GC patients. Its prognostic value was also suggested in both our GC cohort from Zhongshan Hospital and the public databases (KM plotter and GEPIA database). Mechanistically, high NOTCH3 expression correlated with a lower infiltration of activated CD8+ T cells and a higher infiltration of immunosuppressive cells including Tregs and M2 macrophages in the tumor microenvironment. Moreover, high NOTCH3 expression was accompanied by the increased expression of a series of immune checkpoint inhibitors, resulting in a dampened immune response. Interestingly, NOTCH3 expression had a negative association with well-documented predictive biomarkers of immune checkpoint blockade (ICB) immunotherapy, including tumor mutation burden (TMB), gene expression profiling (GEP) score and innate anti-PD-1 resistance (IPRES) signature. CONCLUSION: These findings uncovered a new mechanism by which NOTCH3 participates in the immune tolerance of GC, implying the potential of NOTCH3 as a therapeutic target or predictive marker for GC patients.

15.
Oncologist ; 24(10): 1311-e989, 2019 10.
Article in English | MEDLINE | ID: mdl-31239311

ABSTRACT

LESSONS LEARNED: The NEO-CLASSIC study provided valuable insight for the clinical efficacy and tolerability profiles of perioperative chemotherapy with oxaliplatin and capecitabine, plus gastrectomy, in patients with localized resectable gastric cancer.The study was designed to explore the potential survival benefits of an eight-cycle, perioperative oxaliplatin and capecitabine (XELOX) schedule in the above-mentioned setting and to explore the feasibility of prolonging the cycles of preoperative chemotherapy. The projected endpoint was not met. BACKGROUND: This multicenter, open-label study (NEO-CLASSIC) evaluated the efficacy and safety of oxaliplatin and capecitabine (XELOX), plus D2 gastrectomy, in localized resectable gastric cancer. METHODS: Patients aged 18-75 years with histologically-confirmed gastric adenocarcinoma (stage T2-4a/N+M0) were given eight cycles of XELOX (four preoperatively, four postoperatively). Each 3-week cycle comprised capecitabine 1,000 mg/m2 twice daily on days 1-14 and oxaliplatin 130 mg/m2 on day 1. Curative D2 gastrectomy was scheduled 2-4 weeks after the last preoperative cycle. The primary objective of the study was to determine the objective response rate (ORR) of XELOX in the preoperative setting. Sample size was calculated by assuming that a minimum of 47 cases would be required to increase the ORR by 15% (from 40% to 55%). With an estimated 10% dropout rate, 55 patients would have to be recruited. RESULTS: Fifty-five patients were enrolled, and one was excluded because of screening failure. R0 resections were achieved in 45 of 54 intent-to-treat patients (83.3%), and four patients received R1 resections (Fig. 1). There were no complete responses, 27 (50.0%) partial responses, 22 cases (40.7%) of stable disease, and 4 (7.4%) of progressive disease. The objective response rate was 50.0%. Median follow-up was 52.97 months; 30 patients (55.6%) had disease progression (Table 1), and median progression-free survival was 20.10 (95% confidence interval: 4.31-35.89) months; median overall survival was 30.77 months (95% confidence interval was not yet available) (Fig. 2). Fifty-four patients completed 209 cycles of preoperative chemotherapy; 42 patients received 133 cycles of postoperative chemotherapy (Table 3). The rate of grade 3-4 adverse events was 8.5% (29/342 cycles); the most frequent events were neutropenia (9/342 cycles) and leukopenia (4/342 cycles). CONCLUSION: These findings suggest that combination therapy with capecitabine and oxaliplatin as perioperative chemotherapy, followed by D2 gastrectomy, is effective and safe in late-stage, locally advanced gastric cancer. Although enrollment exceeded the 47 patients required to identify an increase in the ORR by 15% (from 40% to 55%), results did not meet the primary endpoint.


Subject(s)
Adenocarcinoma/drug therapy , Adenocarcinoma/surgery , Capecitabine/therapeutic use , Combined Modality Therapy/methods , Gastrectomy/methods , Oxaliplatin/therapeutic use , Perioperative Period/methods , Stomach Neoplasms/drug therapy , Stomach Neoplasms/surgery , Adenocarcinoma/pathology , Adult , Aged , Capecitabine/pharmacology , Female , Humans , Male , Middle Aged , Oxaliplatin/pharmacology , Stomach Neoplasms/pathology
16.
Biochem Biophys Res Commun ; 512(1): 131-136, 2019 04 23.
Article in English | MEDLINE | ID: mdl-30871776

ABSTRACT

The prognosis of gastric cancer (GC) remains poor due to local invasion and distal metastasis. The GC-related molecular mechanisms underlying invasion and metastasis are not well understood. In this study, we investigated the functional role of ANO1 in GC progression. We found that ANO1 is overexpressed in GC tissues and correlated with GC tumor-node-metastasis stage. Knockdown of ANO1 significantly inhibited GC cell migration and invasion in vitro, and loss of ANO1 resulted in inhibition of tumor metastasis in vivo. Mechanistically, SP1 increased ANO1 transcription, recruited MLL1 to the ANO1 promoter region, facilitated H3K4 trimethylation, and subsequently promoted ANO1 expression. Together, our findings provide a mechanistic assessment of ANO1 overexpression, which represents a GC progression-related molecule and a potentially valuable target for future research.


Subject(s)
Anoctamin-1/genetics , Neoplasm Proteins/genetics , Stomach Neoplasms/genetics , Stomach Neoplasms/metabolism , Animals , Anoctamin-1/antagonists & inhibitors , Binding Sites/genetics , Cell Line, Tumor , Cell Movement , Disease Progression , Female , Gene Knockdown Techniques , Heterografts , Histone-Lysine N-Methyltransferase/genetics , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/secondary , Mice , Mice, Nude , Myeloid-Lymphoid Leukemia Protein/genetics , Myeloid-Lymphoid Leukemia Protein/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis/genetics , Neoplasm Metastasis/pathology , Neoplasm Proteins/antagonists & inhibitors , Promoter Regions, Genetic , Sp1 Transcription Factor/metabolism , Stomach Neoplasms/pathology , Transcriptional Activation
17.
Cancer Manag Res ; 11: 2007-2013, 2019.
Article in English | MEDLINE | ID: mdl-30881125

ABSTRACT

PURPOSE: Chemotherapy is the standard care for patients with incurable advanced gastric cancer. Whether or when the addition of gastrectomy to chemotherapy improves survival of advanced gastric cancer patients with a single noncurable factor remains controversial. We aimed to evaluate the superiority of gastrectomy following chemotherapy vs chemotherapy alone regarding overall survival (OS) in these patients. PATIENTS AND METHODS: Patients with advanced gastric cancer from January 2008 to December 2014 were retrieved from our prospectively acquired database and retrospectively analyzed. The patients with a single noncurable factor were grouped in terms of cancer treatment: chemotherapy alone or gastrectomy following chemotherapy. RESULTS: Four hundred and fourteen patients (333 chemotherapy alone and 81 gastrectomy following chemotherapy) were included in this study. Kaplan-Meier survival curve showed a significant difference on median OS between chemotherapy-alone group and the gastrectomy plus chemotherapy group (10.9 vs 15.9 months, P<0.01). After propensity score analysis (n=126), chemotherapy plus surgery (81 patients) also showed survival benefit over chemotherapy alone (35 patients) (15.9 vs 10.0 months, P<0.01). Furthermore, stratified analyses indicated that patients with liver metastasis, <65 years of age, male, having normal level of carcinoembryonic antigen (CEA) and carbohydrate antigen 19-9 (CA199) upon diagnosis, or having nongastro-esophageal junction tumor benefited from surgery. CONCLUSION: This study suggests that gastrectomy after chemotherapy could lead to survival benefit over chemotherapy alone in advanced gastric cancer patients with a single nonresectable factor if the disease was controllable by chemotherapy.

18.
Gastric Cancer ; 22(3): 587-597, 2019 05.
Article in English | MEDLINE | ID: mdl-30426294

ABSTRACT

BACKGROUND: To compare the efficacy of oxaliplatin-based and oxaliplatin-free adjuvant chemotherapies in patients with different Lauren type gastric cancers after D2 gastrectomy. METHODS: From our established gastric cancer database, patients with pathological stage II and III gastric cancer who received adjuvant chemotherapy after D2 gastrectomy at Zhongshan Hospital of Fudan University were analyzed. Patients who received different adjuvant chemotherapy regimens were divided into two subgroups: oxaliplatin-based and oxaliplatin-free subgroup. Clinical outcomes were analyzed according to pathological stage and different Lauren types. RESULTS: From Jan 2010 to June 2017, a total of 580 patients met all the eligibility criteria and were enrolled. The median DFS for all the patients was 24.37 months and the median OS was 56.70 months. In patients with intestinal type gastric cancer, the median DFS of the oxaliplatin-based subgroup was significantly longer than that of oxaliplatin-free subgroup (48.73 vs. 18.33 months, P < 0.001). The median OS was not reached in the oxaliplatin-based subgroup and 54.33 months in the oxaliplatin-free subgroup (P = 0.006). In patients with diffuse type gastric cancer, neither DFS nor OS differed significantly between two subgroups. In multivariate analysis, oxaliplatin-based adjuvant chemotherapy was independent positive predictor of DFS (HR 0.40; 95% CI 0.28-0.59; P < 0.001) and OS (HR 0.35; 95% CI 0.20-0.62; P < 0.001) in patients with intestinal type gastric cancer. CONCLUSIONS: The results of our study suggested that oxaliplatin-based adjuvant chemotherapy was more effective in patients with intestinal type gastric cancer after D2 gastrectomy but showed no more survival benefit in patients with diffuse type.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Gastrectomy/methods , Stomach Neoplasms/drug therapy , Adenocarcinoma/secondary , Adenocarcinoma/surgery , Capecitabine/administration & dosage , Chemotherapy, Adjuvant , Female , Fluorouracil/administration & dosage , Follow-Up Studies , Humans , Lymphatic Metastasis , Male , Middle Aged , Oxaliplatin/administration & dosage , Postoperative Care , Prognosis , Stomach Neoplasms/pathology , Stomach Neoplasms/surgery , Survival Rate
19.
Onco Targets Ther ; 11: 7061-7075, 2018.
Article in English | MEDLINE | ID: mdl-30410363

ABSTRACT

BACKGROUND: Many studies have discussed the relationship between routine blood parameters and the prognosis of gastric cancer patients; however, few studies focused on the association of routine blood parameters with the efficacy of neoadjuvant chemotherapy (NAC). PATIENTS AND METHODS: We retrospectively collected routine blood parameters and other clinicopathological data of 104 patients with locally advanced gastric cancer (LAGC) who received the oxaliplatin and capecitabine regimen as NAC from June 2010 to March 2016. The objective response rate (ORR), pathological remission rate (pRR), overall survival (OS), and time to recurrence (TTR) were analyzed through different statistical methods, such as Chi-squared test, log-rank test, logistic regression, and Cox regression. RESULTS: In the multivariate analysis, a high platelet-to-lymphocyte ratio (PLR) (≥130.7) predicted a low ORR (OR =5.927, 95% CI: 2.184-16.089) and a low pRR (OR =8.343, 95% CI: 2.178-31.962), while a high lymphocyte-to-white blood cell ratio (LWR) (≥0.228) independently predicted a high ORR (OR =0.118, 95% CI: 0.031-0.448) and a high pRR (OR =0.096, 95% CI: 0.021-0.426). High lymphocyte level (≥1.750×109/L) was an independent predictor of long OS (HR =0.428, 95% CI: 0.190-0.964) and long TTR (HR =0.328, 95% CI: 0.156-0.690). High monocyte level (≥0.215×109/L) was associated with a high pRR (OR =0.072, 95% CI: 0.008-0.636) and a long OS (HR = 0.506, 95% CI: 0.257-0.997). CONCLUSION: In patients with LAGC treated with the oxaliplatin and capecitabine regimen as NAC, a low PLR (<130.7) and a high LWR (≥0.228) independently predicted a high ORR and pRR. High monocyte level (≥0.215×109/L) was an independent predictor for a high pRR and long OS, while patients with high lymphocyte level (≥1.750×109/L) tended to have a long OS and TTR.

20.
BMC Cancer ; 18(1): 702, 2018 Jun 28.
Article in English | MEDLINE | ID: mdl-29954358

ABSTRACT

BACKGROUND: The aim of this study was to compare the efficacies of the XELOX and DOS regimens as preoperative chemotherapy in patients with locally advanced gastric cancer. METHODS: All cases of locally advanced gastric cancer treated with the XELOX or DOS regimen were reviewed retrospectively. Propensity score matching (PSM) was carried out to reduce selection bias based on age, gender, location, Lauren type, carcinoembryonic antigen level, clinical tumor stage, and clinical node stage. RESULTS: From January 2010 to December 2016, 248 patients were matched; 159 of them received the XELOX regimen and 89 the DOS regimen. The response rates in the XELOX and DOS groups were 34.5 and 38.1%, respectively (P = 0.823). After four cycles of chemotherapy, 111 patients (69.8%) in the XELOX group and 65 patients (73.0%) in the DOS group underwent radical surgery (P = 0.485). The median progression-free survival (33.0 months vs. 18.7 months, P = 0.0356) and the median overall survival (43.8 months vs. 29.1 months, P = 0.0003) were longer for patients who received the DOS regimen than for those who received the XELOX regimen. The occurrence of grade 3 to 4 toxicity was similar in the two groups. CONCLUSIONS: For locally advanced gastric cancer patients, the DOS regimen showed more benefit than the XELOX regimen as preoperative chemotherapy, without any added toxicity effects.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Deoxycytidine/analogs & derivatives , Fluorouracil/analogs & derivatives , Propensity Score , Stomach Neoplasms/drug therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Capecitabine , Chemotherapy, Adjuvant , Deoxycytidine/therapeutic use , Docetaxel/administration & dosage , Drug Combinations , Female , Fluorouracil/therapeutic use , Humans , Male , Middle Aged , Oxaliplatin/administration & dosage , Oxaloacetates , Oxonic Acid/administration & dosage , Retrospective Studies , Stomach Neoplasms/mortality , Tegafur/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...