Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 90
Filter
1.
Front Immunol ; 15: 1312380, 2024.
Article in English | MEDLINE | ID: mdl-38726002

ABSTRACT

Objective: The choice of neoadjuvant therapy for esophageal squamous cell carcinoma (ESCC) is controversial. This study aims to provide a basis for clinical treatment selection by establishing a predictive model for the efficacy of neoadjuvant immunochemotherapy (NICT). Methods: A retrospective analysis of 30 patients was conducted, divided into Response and Non-response groups based on whether they achieved major pathological remission (MPR). Differences in genes and immune microenvironment between the two groups were analyzed through next-generation sequencing (NGS) and multiplex immunofluorescence (mIF). Variables most closely related to therapeutic efficacy were selected through LASSO regression and ROC curves to establish a predictive model. An additional 48 patients were prospectively collected as a validation set to verify the model's effectiveness. Results: NGS suggested seven differential genes (ATM, ATR, BIVM-ERCC5, MAP3K1, PRG, RBM10, and TSHR) between the two groups (P < 0.05). mIF indicated significant differences in the quantity and location of CD3+, PD-L1+, CD3+PD-L1+, CD4+PD-1+, CD4+LAG-3+, CD8+LAG-3+, LAG-3+ between the two groups before treatment (P < 0.05). Dynamic mIF analysis also indicated that CD3+, CD8+, and CD20+ all increased after treatment in both groups, with a more significant increase in CD8+ and CD20+ in the Response group (P < 0.05), and a more significant decrease in PD-L1+ (P < 0.05). The three variables most closely related to therapeutic efficacy were selected through LASSO regression and ROC curves: Tumor area PD-L1+ (AUC= 0.881), CD3+PD-L1+ (AUC= 0.833), and CD3+ (AUC= 0.826), and a predictive model was established. The model showed high performance in both the training set (AUC= 0.938) and the validation set (AUC= 0.832). Compared to the traditional CPS scoring criteria, the model showed significant improvements in accuracy (83.3% vs 70.8%), sensitivity (0.625 vs 0.312), and specificity (0.937 vs 0.906). Conclusion: NICT treatment may exert anti-tumor effects by enriching immune cells and activating exhausted T cells. Tumor area CD3+, PD-L1+, and CD3+PD-L1+ are closely related to therapeutic efficacy. The model containing these three variables can accurately predict treatment outcomes, providing a reliable basis for the selection of neoadjuvant treatment plans.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Neoadjuvant Therapy , Tumor Microenvironment , Humans , Tumor Microenvironment/immunology , Esophageal Squamous Cell Carcinoma/therapy , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/drug therapy , Neoadjuvant Therapy/methods , Esophageal Neoplasms/therapy , Esophageal Neoplasms/immunology , Esophageal Neoplasms/drug therapy , Male , Female , Middle Aged , Retrospective Studies , Prognosis , Aged , Biomarkers, Tumor , Treatment Outcome , Immunotherapy/methods
2.
J Immunother Cancer ; 12(5)2024 May 09.
Article in English | MEDLINE | ID: mdl-38724465

ABSTRACT

BACKGROUND: CD276 (B7-H3), a pivotal immune checkpoint, facilitates tumorigenicity, invasiveness, and metastasis by escaping immune surveillance in a variety of tumors; however, the underlying mechanisms facilitating immune escape in esophageal squamous cell carcinoma (ESCC) remain enigmatic. METHODS: We investigated the expression of CD276 in ESCC tissues from patients by using immunohistochemistry (IHC) assays. In vivo, we established a 4-nitroquinoline 1-oxide (4NQO)-induced CD276 knockout (CD276wKO) and K14cre; CD276 conditional knockout (CD276cKO) mouse model of ESCC to study the functional role of CD276 in ESCC. Furthermore, we used the 4NQO-induced mouse model to evaluate the effects of anti-CXCL1 antibodies, anti-Ly6G antibodies, anti-NK1.1 antibodies, and GSK484 inhibitors on tumor growth. Moreover, IHC, flow cytometry, and immunofluorescence techniques were employed to measure immune cell proportions in ESCC. In addition, we conducted single-cell RNA sequencing analysis to examine the alterations in tumor microenvironment following CD276 depletion. RESULTS: In this study, we elucidate that CD276 is markedly upregulated in ESCC, correlating with poor prognosis. In vivo, our results indicate that depletion of CD276 inhibits tumorigenesis and progression of ESCC. Furthermore, conditional knockout of CD276 in epithelial cells engenders a significant downregulation of CXCL1, consequently reducing the formation of neutrophil extracellular trap networks (NETs) via the CXCL1-CXCR2 signaling axis, while simultaneously augmenting natural killer (NK) cells. In addition, overexpression of CD276 promotes tumorigenesis via increasing NETs' formation and reducing NK cells in vivo. CONCLUSIONS: This study successfully elucidates the functional role of CD276 in ESCC. Our comprehensive analysis uncovers the significant role of CD276 in modulating immune surveillance mechanisms in ESCC, thereby suggesting that targeting CD276 might serve as a potential therapeutic approach for ESCC treatment.


Subject(s)
B7 Antigens , Chemokine CXCL1 , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Receptors, Interleukin-8B , Animals , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Mice , Humans , Receptors, Interleukin-8B/metabolism , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , B7 Antigens/metabolism , Chemokine CXCL1/metabolism , Extracellular Traps/metabolism , Tumor Escape , Female , Male , Mice, Knockout , Tumor Microenvironment
3.
Cancer Med ; 13(9): e7228, 2024 May.
Article in English | MEDLINE | ID: mdl-38733174

ABSTRACT

BACKGROUND: The molecular and immunological characteristics of primary tumors and positive lymph nodes in esophageal squamous cell carcinoma (ESCC) are unknown and the relationship with recurrence is unclear, which this study attempted to explore. METHODS: A total of 30 ESCC patients with lymph node positive (IIB-IVA) were enrolled. Among them, primary tumor and lymph node specimens were collected from each patient, and subjected to 551-tumor-targeted DNA sequencing and 289-immuno-oncology RNA panel sequencing to identify the different molecular basis and immunological features, respectively. RESULTS: The primary tumors exhibited a higher mutation burden than lymph nodes (p < 0.001). One-year recurrent ESCC exhibited a higher Mucin16 (MUC16) mutation rate (p = 0.038), as well as univariate and multivariate analysis revealed that MUC16 mutation is independent genetic factor associated with reduced relapse-free survival (univariate, HR: 5.39, 95% CI: 1.67-17.4, p = 0.005; multivariate, HR: 7.36, 95% CI: 1.79-30.23, p = 0.006). Transcriptomic results showed non-relapse group had higher cytolytic activity (CYT) score (p = 0.025), and was enriched in the IFN-α pathway (p = 0.036), while those in the relapsed group were enriched in the TNF-α/NF-κB (p = 0.001) and PI3K/Akt pathway (p = 0.014). CONCLUSION: The difference in molecular characteristics between primary lesions and lymph nodes may be the cause of the inconsistent clinical outcomes. Mutations of MUC16 and poor immune infiltration are associated with rapid relapse of nodes-positive ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lymph Nodes , Lymphatic Metastasis , Mutation , Neoplasm Recurrence, Local , Humans , Male , Female , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/surgery , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Middle Aged , Neoplasm Recurrence, Local/genetics , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophageal Neoplasms/mortality , Lymph Nodes/pathology , Lymph Nodes/immunology , Aged , Biomarkers, Tumor/genetics , Prognosis , Membrane Proteins , CA-125 Antigen
4.
Cancer Invest ; 42(3): 243-259, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38616306

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) presents a five-year survival rate below 20%, underscoring the need for improved prognostic markers. Our study analyzed ESCC-specific datasets to identify consistently differentially expressed genes. A Venn analysis followed by gene network interactions revealed 23 key genes, from which we built a prognostic model using the COX algorithm (p = 0.000245, 3-year AUC = 0.967). This model stratifies patients into risk groups, with high-risk individuals showing worse outcomes and lower chemotherapy sensitivity. Moreover, a link between risk scores and M2 macrophage infiltration, as well as significant correlations with immune checkpoint genes (e.g., SIGLEC15, PDCD1LG2, and HVCR2), was discovered. High-risk patients had lower Tumor Immune Dysfunction and Exclusion (TIDE) values, suggesting potential responsiveness to immune checkpoint blockade (ICB) therapy. Our efficient 23-gene prognostic model for ESCC indicates a dual utility in assessing prognosis and guiding therapeutic decisions, particularly in the context of ICB therapy for high-risk patients.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/mortality , Prognosis , Esophageal Neoplasms/genetics , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/immunology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Biomarkers, Tumor/genetics , Male , Female , Gene Expression Regulation, Neoplastic , Drug Resistance, Neoplasm/genetics , Middle Aged , Gene Regulatory Networks
5.
J Gene Med ; 26(3): e3667, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38442944

ABSTRACT

BACKGROUND: Esophageal squamous cell carcinoma (ESCC) is a predominant subtype of esophageal cancer with relatively high mortality worldwide. Serine peptidase inhibitor Kazal-type 5 (SPINK5) is reported to be downregulated in ESCC. However, its explicit role in ESCC remains further investigation. METHODS: The tumor tissues and adjacent non-cancerous tissues were obtained from 196 patients with ESCC for the determination of SPINK5 mRNA levels. Additionally, the relationship between SPINK5 mRNA levels and clinicopathological features of ESCC patients was explored. The effects of SPINK5 on the invasion and migration of ESCC cells were assessed using Transwell assays. Furthermore, SPINK5 mRNA and LEKTI protein were measured in ESCC cell lines after treatment with poly (I:C), lipopolysaccharide (LPS) or unmethylated CpG DNA. Moreover, the correlation between expression of SPINK5 and nuclear factor-kappa B (NF-κB) signaling pathway-related genes was analyzed in the TCGA-ESCC cohort, and the effects of SPINK5 on NF-κB transcription was analyzed using a luciferase reporter gene assay. Finally, the correlations between SPINK5 and infiltration of immune cells, immune scores, stromal scores and ESTIMATE (i.e., Estimation of STromal and Immune cells in MAlignant Tumor tissues using Expression data) scores were explored. RESULTS: SPINK5 mRNA levels were downregulated in tumor tissues, which was significantly correlated with higher lymph node metastases. Overexpressed SPINK5 inhibited cell invasion and migration in ESCC cell lines. Mechanistically, LPS-induced activation of Toll-like receptor 4 (TLR4) decreased SPINK5 mRNA and LEKTI in KYSE150 and KYSE70 cells. Spearman correlation analysis revealed that SPINK5 mRNA was significantly negatively correlated with a total of seven NF-κB signaling pathway-related genes in TCGA-ESCC patients. Moreover, downregulation of SPINK5 increased and upregulation of SPINK5 decreased the activity of the NF-κB promoter in HEK293T cells. Finally, immune cells infiltration analysis revealed that SPINK5 was significantly correlated with the infiltration of various immune cells, stromal scores, immune scores and ESTIMATE scores. CONCLUSIONS: SPINK5 plays critical roles in the TLR4/NF-κB pathway and immune cells infiltration, which might contribute to the ESCC metastasis. The findings of the present study may provide a promising biomarker for the diagnosis and treatment of esophageal squamous cell carcinoma.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Serine Peptidase Inhibitor Kazal-Type 5 , Humans , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/metabolism , HEK293 Cells , Lipopolysaccharides , NF-kappa B/metabolism , RNA, Messenger/metabolism , Serine Peptidase Inhibitor Kazal-Type 5/metabolism , Toll-Like Receptor 4/metabolism
6.
Biomol Biomed ; 24(3): 545-559, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38340316

ABSTRACT

The enzyme glutamate-cysteine ligase modifier subunit (GCLM) serves as the initial rate-limiting factor in glutathione (GSH) synthesis. GSH is the preferred substrate for glutathione peroxidase 4 (GPX4), directly impacting its activity and stability. This study aims to elucidate the expression of GCLM and its correlation with the nuclear factor erythroid 2-related factor 2 (NFE2L2), commonly referred to as NRF2, in esophageal squamous cell carcinoma (ESCC) and further investigate the potential signaling axis of radiotherapy resistance caused by NRF2-mediated regulation of ferroptosis in ESCC. The expression of NRF2, GCLM, and GPX4 in ESCC was analyzed by bioinformatics, and their relationship with ferroptosis was verified through cell function experiments. Their role in radioresistance was then investigated through multiple validation steps. Bioinformatics analysis was employed to determine the immune infiltration pattern of NRF2 in ESCC. Furthermore, the effect of NRF2-mediated massive macrophage M2 infiltration on radiotherapy and ferroptosis was validated through in vivo experiments. In vitro assays demonstrated that overactivated NRF2 promotes radioresistance by directly binding to the promoter region of GCLM. The Tumor Immune Estimation Resource (TIMER) and quanTIseq analyses revealed NRF2 enrichment in M2 macrophages with a positive correlation. Co-culturing KYSE450 cells with M2 macrophages demonstrated that a significant infiltration of macrophages M2 can render ESCC cells resistant to radiotherapy but restore their sensitivity to ferroptosis in the process. Our study elucidates a link between the NRF2-GCLM-GSH-GPX4 signaling axis in ESCC, highlighting its potential as a therapeutic target for antagonistic biomarkers of resistance in the future. Additionally, it provides a novel treatment avenue for ESCC metastasis and radioresistance.


Subject(s)
Ferroptosis , Glutamate-Cysteine Ligase , NF-E2-Related Factor 2 , Radiation Tolerance , Humans , NF-E2-Related Factor 2/metabolism , NF-E2-Related Factor 2/genetics , Glutamate-Cysteine Ligase/metabolism , Glutamate-Cysteine Ligase/genetics , Radiation Tolerance/genetics , Cell Line, Tumor , Animals , Mice , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/metabolism , Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/radiotherapy , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/immunology , Gene Expression Regulation, Neoplastic , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Phospholipid Hydroperoxide Glutathione Peroxidase/genetics , Mice, Nude , Male , Mice, Inbred BALB C
7.
Adv Healthc Mater ; 13(11): e2303623, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38142309

ABSTRACT

PD-1/PD-L1 blockade immunotherapy has gained approval for the treatment of a diverse range of tumors; however, its efficacy is constrained by the insufficient infiltration of T lymphocytes into the tumor microenvironment, resulting in suboptimal patient responses. Here, a pioneering immunotherapy ferritin nanodrug delivery system denoted as ITFn-Pt(IV) is introduced. This system orchestrates a synergistic fusion of PD-L1 blockade, chemotherapy, and T-cell activation, aiming to augment the efficacy of tumor immunotherapy. Leveraging genetic engineering approach and temperature-regulated channel-based drug loading techniques, the architecture of this intelligent responsive system is refined. It is adept at facilitating the precise release of T-cell activating peptide Tα1 in the tumor milieu, leading to an elevation in T-cell proliferation and activation. The integration of PD-L1 nanobody KN035 ensures targeted engagement with tumor cells and mediates the intracellular delivery of the encapsulated Pt(IV) drugs, culminating in immunogenic cell death and the subsequent dendritic cell maturation. Employing esophageal squamous cell carcinoma (ESCC) as tumor model, the potent antitumor efficacy of ITFn-Pt(IV) is elucidated, underscored by augmented T-cell infiltration devoid of systemic adverse effects. These findings accentuate the potential of ITFn-Pt(IV) for ESCC treatment and its applicability to other malignancies resistant to established PD-1/PD-L1 blockade therapies.


Subject(s)
B7-H1 Antigen , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , T-Lymphocytes , Animals , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/pathology , Esophageal Neoplasms/immunology , Humans , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/immunology , B7-H1 Antigen/metabolism , Mice , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/therapy , Cell Line, Tumor , Lymphocyte Activation/drug effects , Ferritins/chemistry , Tumor Microenvironment/drug effects , Immunotherapy/methods , Antineoplastic Agents/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/therapeutic use , Nanoparticles/chemistry , Drug Delivery Systems/methods
8.
J Immunother Cancer ; 10(4)2022 04.
Article in English | MEDLINE | ID: mdl-35428705

ABSTRACT

BACKGROUND: Type 1 conventional dendritic cells (cDC1s) are characterized by their ability to induce potent CD8+ T cell responses. In efforts to generate novel vaccination strategies, notably against cancer, human cDC1s emerge as an ideal target to deliver antigens. cDC1s uniquely express XCR1, a seven transmembrane G protein-coupled receptor. Due to its restricted expression and endocytic nature, XCR1 represents an attractive receptor to mediate antigen-delivery to human cDC1s. METHODS: To explore tumor antigen delivery to human cDC1s, we used an engineered version of XCR1-binding lymphotactin (XCL1), XCL1(CC3). Site-specific sortase-mediated transpeptidation was performed to conjugate XCL1(CC3) to an analog of the HLA-A*02:01 epitope of the cancer testis antigen New York Esophageal Squamous Cell Carcinoma-1 (NY-ESO-1). While poor epitope solubility prevented isolation of stable XCL1-antigen conjugates, incorporation of a single polyethylene glycol (PEG) chain upstream of the epitope-containing peptide enabled generation of soluble XCL1(CC3)-antigen fusion constructs. Binding and chemotactic characteristics of the XCL1-antigen conjugate, as well as its ability to induce antigen-specific CD8+ T cell activation by cDC1s, was assessed. RESULTS: PEGylated XCL1(CC3)-antigen conjugates retained binding to XCR1, and induced cDC1 chemoattraction in vitro. The model epitope was efficiently cross-presented by human cDC1s to activate NY-ESO-1-specific CD8+ T cells. Importantly, vaccine activity was increased by targeting XCR1 at the surface of cDC1s. CONCLUSION: Our results present a novel strategy for the generation of targeted vaccines fused to insoluble antigens. Moreover, our data emphasize the potential of targeting XCR1 at the surface of primary human cDC1s to induce potent CD8+ T cell responses.


Subject(s)
Antigens, Neoplasm , Cancer Vaccines , Dendritic Cells , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lymphokines , Membrane Proteins , Sialoglycoproteins , Antigens, Neoplasm/administration & dosage , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Cross-Priming , Dendritic Cells/immunology , Epitopes/immunology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/therapy , Humans , Lymphokines/administration & dosage , Lymphokines/immunology , Male , Membrane Proteins/administration & dosage , Membrane Proteins/immunology , Sialoglycoproteins/administration & dosage , Sialoglycoproteins/immunology
9.
Biomed Res Int ; 2022: 8636527, 2022.
Article in English | MEDLINE | ID: mdl-35463992

ABSTRACT

This study was aimed at exploring the mechanism of serine threonine protein kinase 11 (STK11)/Adenosine 5'-monophosphate-activated protein kinase (AMPK) signaling pathway after immunotherapy for esophageal squamous cell carcinoma (ESCC), providing basic information for the clinical treatment of ESCC. In this study, tissue specimens from 100 patients with ESCC who underwent surgical treatment in Taizhou People's Hospital (group A) and 20 patients with recurrent or metastatic ESCC who received second-line immunotherapy (group B) were collected. The real-time fluorescent quantitative polymerase chain reaction (PCR) (RT-qPCR) technology was used to detect the expression levels of STK11, interferon-γ (IFN-γ), interleukin 6 (IL-6), and vascular endothelial growth factor (VEGF) in the tissues. The immunohistochemical staining was used to detect the positive expression levels (PELs) of STK11 and AMPKα in the tissues, and immunofluorescence staining was used to detect the PELs Teff cells (CD3 and CD8), Treg cells (CD4 and FOXP3), and neutrophils (CD68 and CD163). RT-qPCR results showed that the expression levels of STK11 and IFN-γ in group A were obviously lower, and those of IL-6 and VEGF were much higher in contrast to group B (P < 0.05). The results of immunohistochemical staining showed that the number of STK11- and AMPKα-positive staining cells in group A was dramatically less than that in group B (P <0.05). The results of immunofluorescence staining revealed that the number of positive staining cells for Teff cells, Treg cells, and neutrophils in group A was also less dramatically than that in group B (P <0.05). In summary, immunotherapy can play a therapeutic effect on ESCC by regulating STK11/AMPK pathway and immune cell infiltration.


Subject(s)
AMP-Activated Protein Kinase Kinases , AMP-Activated Protein Kinases , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , AMP-Activated Protein Kinase Kinases/genetics , AMP-Activated Protein Kinase Kinases/immunology , AMP-Activated Protein Kinases/genetics , AMP-Activated Protein Kinases/immunology , Cell Line, Tumor , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/surgery , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/surgery , Esophageal Squamous Cell Carcinoma/therapy , Gene Expression Regulation, Neoplastic , Humans , Immunotherapy/methods , Interleukin-6/immunology , Protein Serine-Threonine Kinases/genetics , Signal Transduction , Vascular Endothelial Growth Factor A/immunology
10.
Anticancer Res ; 42(3): 1499-1507, 2022 Mar.
Article in English | MEDLINE | ID: mdl-35220245

ABSTRACT

BACKGROUND: Trimodal therapy is frequently used for patients with locally advanced, resectable oesophageal cancer. However, it does not provide a satisfactory prognosis. The neutrophil-to-lymphocyte ratio (NLR) is an important indicator of patients' inflammatory and immune statuses. We investigated the prognostic role of NLR values obtained at different treatment stages in patients with oesophageal squamous cell carcinoma. PATIENTS AND METHODS: We evaluated the correlation between NLR values or their change and prognosis at each treatment point (before chemoradiotherapy; before surgery; and at 14 days, and 1 and 2 months postoperatively) in 163 patients with oesophageal squamous cell carcinoma who underwent oesophagectomy after neoadjuvant chemoradiotherapy from April 2003 to August 2018. The outcomes studied were overall (OS) and relapse-free (RFS) survival. RESULTS: The NLR at 1 month postoperatively showed the strongest correlation with prognosis, with an optimal cut-off value of 4.5 (area under the curve=0.7878; 95% confidence interval=0.70-0.85; p<0.0001). Univariate and multivariate analyses showed that NLR ≥4.5 was a significant factor for both RFS (hazard ratio=4.44, 95% confidence interval=2.69-7.34) and OS (hazard ratio=3.88, 95% confidence interval=2.38-6.32). Furthermore, NLR significantly stratified patients for the RFS and OS regardless of the pathological response (complete/non-complete response) and postoperative complications (Clavien-Dindo grade

Subject(s)
Chemoradiotherapy, Adjuvant , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/surgery , Esophagectomy , Lymphocytes/immunology , Neoadjuvant Therapy , Neoplasm Recurrence, Local , Neutrophils/immunology , Aged , Chemoradiotherapy, Adjuvant/adverse effects , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Esophagectomy/adverse effects , Female , Humans , Lymphocyte Count , Male , Middle Aged , Neoadjuvant Therapy/adverse effects , Predictive Value of Tests , Progression-Free Survival , Retrospective Studies , Risk Assessment , Risk Factors , Time Factors
11.
Eur J Immunol ; 52(2): 338-351, 2022 02.
Article in English | MEDLINE | ID: mdl-34755333

ABSTRACT

PSMA3, a member of the proteasome subunit, has been shown to play a major player in protein degradation. Reportedly, PSMA3 functions as a negative regulator in various cancers including colon, pancreatic and gastric cancers. However, the contributions of PSMA3 to the progression of esophageal squamous cell carcinoma (ESCC) and the underlying mechanism remain unclear. Therefore, in this study, we investigated whether PSMA3 is involved in ESCC progression and the potential underlying mechanism. The results revealed that PSMA3 was highly expressed in the ESCC tumor tissues and functioned as a negative indicator according to the data from The Cancer Genome Atlas (TCGA)/Gene Expression Omnibus (GEO) datasets and clinical patients' samples. Pathway enrichment analysis showed that PSMA3 was closely correlated with ESCC cancer stemness and the inflammatory response; however, this correlation was absent after knockdown of PSMA3 in vitro. We further demonstrated that PSMA3 suppressed CD8+ T-cells infiltration depending on the C-C motif chemokine ligand 3 (CCL3)/C-C motif chemokine receptor 5 (CCR5) axis. Collectively, these results demonstrate the role of PSMA3 in ESCC cancer stemness and the negative regulation of CD8 T-cells infiltration mediated by PSMA3. The results of this study may provide a potential target for the immuno-oncology effect of PSMA3 in ESCC therapy.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Gene Expression Regulation, Enzymologic/immunology , Gene Expression Regulation, Neoplastic/immunology , Neoplasm Proteins , Cell Line, Tumor , Databases, Nucleic Acid , Esophageal Neoplasms/enzymology , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Squamous Cell Carcinoma/enzymology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/immunology , Humans , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Proteasome Endopeptidase Complex/biosynthesis , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/immunology
12.
Nat Commun ; 12(1): 7335, 2021 12 17.
Article in English | MEDLINE | ID: mdl-34921160

ABSTRACT

The tumor microenvironment is a highly complex ecosystem of diverse cell types, which shape cancer biology and impact the responsiveness to therapy. Here, we analyze the microenvironment of esophageal squamous cell carcinoma (ESCC) using single-cell transcriptome sequencing in 62,161 cells from blood, adjacent nonmalignant and matched tumor samples from 11 ESCC patients. We uncover heterogeneity in most cell types of the ESCC stroma, particularly in the fibroblast and immune cell compartments. We identify a tumor-specific subset of CST1+ myofibroblasts with prognostic values and potential biological significance. CST1+ myofibroblasts are also highly tumor-specific in other cancer types. Additionally, a subset of antigen-presenting fibroblasts is revealed and validated. Analyses of myeloid and T lymphoid lineages highlight the immunosuppressive nature of the ESCC microenvironment, and identify cancer-specific expression of immune checkpoint inhibitors. This work establishes a rich resource of stromal cell types of the ESCC microenvironment for further understanding of ESCC biology.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression Profiling , Single-Cell Analysis , Tumor Microenvironment/genetics , Antigen Presentation , Biomarkers, Tumor/metabolism , Dendritic Cells/metabolism , Esophageal Neoplasms/immunology , Esophageal Squamous Cell Carcinoma/immunology , Gene Expression Regulation, Neoplastic , Histocompatibility Antigens Class II/metabolism , Humans , Myeloid Cells/metabolism , Myofibroblasts/pathology , Prognosis , Salivary Cystatins/metabolism , Survival Analysis , T-Lymphocytes/metabolism , Tumor Microenvironment/immunology
13.
Int J Mol Sci ; 22(21)2021 Oct 25.
Article in English | MEDLINE | ID: mdl-34768915

ABSTRACT

Ursolic acid (UA), a pentacyclic triterpenoid extracted from various plants, inhibits cell growth, metastasis, and tumorigenesis in various cancers. Chemotherapy resistance and the side effects of paclitaxel (PTX), a traditional chemotherapy reagent, have limited the curative effect of PTX in esophageal cancer. In this study, we investigate whether UA promotes the anti-tumor effect of PTX and explore the underlying mechanism of their combined effect in esophageal squamous cell carcinoma (ESCC). Combination treatment with UA and PTX inhibited cell proliferation and cell growth more effectively than either treatment alone by inducing more significant apoptosis, as indicated by increased sub-G1 phase distribution and protein levels of cleaved-PARP and cleaved caspase-9. Similar to the cell growth suppressive effect, the combination of UA and PTX significantly inhibited cell migration by targeting uPA, MMP-9, and E-cadherin in ESCC cells. In addition, combination treatment with UA and PTX significantly activated p-GSK-3ß and suppressed the activation of Akt and FOXM1 in ESCC cells. Those effects were enhanced by the Akt inhibitor LY2940002 and inverted by the Akt agonist SC79. In an in vivo evaluation of a murine xenograft model of esophageal cancer, combination treatment with UA and PTX suppressed tumor growth significantly better than UA or PTX treatment alone. Thus, UA effectively potentiates the anti-tumor efficacy of PTX by targeting the Akt/FOXM1 cascade since combination treatment shows significantly more anti-tumor potential than PTX alone both in vitro and in vivo. Combination treatment with UA and PTX could be a new strategy for curing esophageal cancer patients.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Esophageal Neoplasms/drug therapy , Esophageal Squamous Cell Carcinoma/drug therapy , Paclitaxel/pharmacology , Triterpenes/pharmacology , Animals , Apoptosis/drug effects , Cadherins/metabolism , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Forkhead Box Protein M1/metabolism , Glycogen Synthase Kinase 3 beta/metabolism , Humans , Matrix Metalloproteinase 9/metabolism , Mice , Neoplasm Invasiveness/pathology , Proto-Oncogene Proteins c-akt/metabolism , Signal Transduction/drug effects , Ursolic Acid
14.
Pathol Res Pract ; 228: 153683, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34798484

ABSTRACT

Chemokines have distinct effects on tumor progression by affecting cancer immunity and tumorigenesis. However, the characteristic chemokine profiles and their roles in immune cell recruitment and cancer cell biology are not entirely understood in esophageal cancer. Here, we scrutinized chemokine's expression profiles in independent esophageal cancer cohorts and identified the elevated CCL20 as a risk factor to predict patients' prognosis regardless of histology subtypes. Enhanced CCL20 expression was also associated with the acquisition of metastatic potential. Mechanistically, the upregulation of CCL20 in tumor cells was associated with promoter hypomethylation. Furthermore, by analyzing single-cell RNA sequencing data of a mouse model mimicking human ESCC development, we observed an imbalance among CD4+ T subtypes in the tumor microenvironment, namely Ccr6+ Th17 and Treg cells infiltration alongside the elevated Ccl20 expression in abnormal epithelial cells during the tumorigenic process. Together, these results reveal that hypomethylation-induced CCL20 promotes esophageal cancer progression and immune disorder. Targeting CCL20 might be a promising therapeutic approach in esophageal cancer.


Subject(s)
Chemokine CCL20/biosynthesis , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/pathology , Animals , Chemokine CCL20/genetics , Cohort Studies , DNA Methylation , Disease Progression , Gene Expression Regulation/physiology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Tumor Microenvironment/immunology , Up-Regulation
15.
Biomed Res Int ; 2021: 7430315, 2021.
Article in English | MEDLINE | ID: mdl-34722771

ABSTRACT

Immune checkpoint inhibitor (ICI) therapy may benefit patients with advanced esophageal squamous cell carcinoma (ESCC); however, novel biomarkers are needed to help predict the response of patients to treatment. Differentially expressed immune-related genes within The Cancer Genome Atlas ESCC dataset were selected using the weighted gene coexpression network and lasso Cox regression analyses. Based on these data, an immune-related gene prognostic index (IRGPI) was constructed. The molecular characteristics of the different IRGPI subgroups were assessed using mutation information and gene set enrichment analysis. Differences in immune cell infiltration and the response to ICI therapy and other drugs were also analyzed. Additionally, tumor and adjacent control tissues were collected from six patients with ESCC and the expression of these genes was verified using real-time quantitative polymerase chain reaction. IRGPI was designed based on CLDN1, HCAR3, FNBP1L, and BRCA2, the expression of which was confirmed in ESCC samples. The prognosis of patients in the high-IRGPI group was poor, as verified using publicly available expression data. KMT2D mutations were more common in the high-IRGPI group. Enrichment analysis revealed an active immune response, and immune infiltration assessment showed that the high-IRGPI group had an increased infiltration degree of CD8 T cells, which contributed to the improved response to ICI treatment. Collectively, these data demonstrate that IRGPI is a robust biomarker for predicting the prognosis and response to therapy of patients with ESCC.


Subject(s)
Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/immunology , BRCA2 Protein/genetics , Biomarkers, Pharmacological , Carrier Proteins/genetics , Claudin-1/genetics , Computational Biology/methods , Databases, Genetic , Esophageal Neoplasms/genetics , Esophageal Neoplasms/immunology , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Gene Expression/genetics , Gene Expression Profiling/methods , Gene Expression Regulation, Neoplastic/genetics , Gene Regulatory Networks/genetics , Humans , Prognosis , Receptors, G-Protein-Coupled/genetics , Receptors, Nicotinic/genetics , Transcriptome/genetics
16.
Biomed Res Int ; 2021: 8635381, 2021.
Article in English | MEDLINE | ID: mdl-34616847

ABSTRACT

Tumor-infiltrating immune cells have been implicated in the tumorigenesis and progression of esophageal squamous cell carcinoma (ESCC). However, the functionalities and clinical significance of immune cells remain largely unveiled. In this study, the gene expression data from the Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) were extracted. The relative infiltrating levels were estimated by single-sample gene set enrichment analysis. Some cytotoxic immune cells were attenuated, and resting cytotoxic immune cells were accumulated in ESCC. Remarkably, we also observed that infiltrating levels of macrophage M2 and resting natural killer (NK) cells were increased in nonresponders of CRT, and T cells that had anticancer activities such as activated memory CD4 and T helper 2 (Th2) cells were significantly reduced in ESCC tissues of the nonresponders. Moreover, the high infiltrations of the resting natural killer (NK) and dendritic cell (DC) were observed to result in a shorter overall survival in ESCC. Consistently, high expression of immune checkpoint genes, CTLA4 and HAVCR2, was associated with poor prognosis. Furthermore, STAT5B, a key transcription factor, as well as its target genes, involved in the regulation of T cells, was significantly downregulated in ESCC, especially subgroup I, indicating that downregulation of STAT5B might be associated with reduced T cell-mediated anticancer activity. In conclusion, the present study significantly improved our understanding of the regulatory roles of immune cells in ESCC.


Subject(s)
Esophageal Neoplasms/immunology , Esophageal Squamous Cell Carcinoma/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Down-Regulation/genetics , Esophageal Neoplasms/drug therapy , Esophageal Neoplasms/genetics , Esophageal Neoplasms/radiotherapy , Esophageal Squamous Cell Carcinoma/drug therapy , Esophageal Squamous Cell Carcinoma/genetics , Esophageal Squamous Cell Carcinoma/radiotherapy , Gene Expression Regulation, Neoplastic , Humans , Immune Checkpoint Inhibitors
17.
Nat Commun ; 12(1): 5291, 2021 09 06.
Article in English | MEDLINE | ID: mdl-34489433

ABSTRACT

Esophageal squamous-cell carcinoma (ESCC), one of the most prevalent and lethal malignant disease, has a complex but unknown tumor ecosystem. Here, we investigate the composition of ESCC tumors based on 208,659 single-cell transcriptomes derived from 60 individuals. We identify 8 common expression programs from malignant epithelial cells and discover 42 cell types, including 26 immune cell and 16 nonimmune stromal cell subtypes in the tumor microenvironment (TME), and analyse the interactions between cancer cells and other cells and the interactions among different cell types in the TME. Moreover, we link the cancer cell transcriptomes to the somatic mutations and identify several markers significantly associated with patients' survival, which may be relevant to precision care of ESCC patients. These results reveal the immunosuppressive status in the ESCC TME and further our understanding of ESCC.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Neoplasm Proteins/genetics , Stromal Cells/immunology , Transcription, Genetic , Adult , Aged , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Epithelial Cells/immunology , Epithelial Cells/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Female , Fibroblasts/immunology , Fibroblasts/pathology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Male , Middle Aged , Myeloid Cells/immunology , Myeloid Cells/pathology , Neoplasm Proteins/classification , Neoplasm Proteins/immunology , Prognosis , Single-Cell Analysis , Stromal Cells/pathology , Survival Analysis , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Whole Genome Sequencing
18.
Sci Rep ; 11(1): 16696, 2021 08 17.
Article in English | MEDLINE | ID: mdl-34404882

ABSTRACT

Esophageal squamous cell carcinoma (ESCC) is a life-threatening thoracic tumor with a poor prognosis. The role of molecular alterations and the immune microenvironment in ESCC development has not been fully elucidated. The present study aimed to elucidate key candidate genes and immune cell infiltration characteristics in ESCC by integrated bioinformatics analysis. Nine gene expression datasets from the Gene Expression Omnibus (GEO) database were analysed to identify robust differentially expressed genes (DEGs) using the robust rank aggregation (RRA) algorithm. Functional enrichment analyses showed that the 152 robust DEGs are involved in multiple processes in the tumor microenvironment (TME). Immune cell infiltration analysis based on the 9 normalized GEO microarray datasets was conducted with the CIBERSORT algorithm. The changes in macrophages between ESCC and normal tissues were particularly obvious. In ESCC tissues, M0 and M1 macrophages were increased dramatically, while M2 macrophages were decreased. A robust DEG-based protein-protein interaction (PPI) network was used for hub gene selection with the CytoHubba plugin in Cytoscape. Nine hub genes (CDA, CXCL1, IGFBP3, MMP3, MMP11, PLAU, SERPINE1, SPP1 and VCAN) had high diagnostic efficiency for ESCC according to receiver operating characteristic (ROC) curve analysis. The expression of all hub genes except MMP3 and PLAU was significantly related to macrophage infiltration. Univariate and multivariate regression analyses showed that a 7-gene signature constructed from the robust DEGs was useful for predicting ESCC prognosis. Our results might facilitate the exploration of potential targeted TME therapies and prognostic evaluation in ESCC.


Subject(s)
Esophageal Neoplasms/genetics , Esophageal Squamous Cell Carcinoma/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Esophageal Neoplasms/immunology , Esophageal Squamous Cell Carcinoma/diagnosis , Esophageal Squamous Cell Carcinoma/immunology , Gene Regulatory Networks , Humans , Immunity, Cellular , Prognosis , Tumor Microenvironment
19.
J Clin Oncol ; 39(28): 3182-3184, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34406872

ABSTRACT

ASCO Rapid Recommendations Updates highlight revisions to select ASCO guideline recommendations as a response to the emergence of new and practice-changing data. The rapid updates are supported by an evidence review and follow the guideline development processes outlined in the ASCO Guideline Methodology Manual. The goal of these articles is to disseminate updated recommendations, in a timely manner, to better inform health practitioners and the public on the best available cancer care options.


Subject(s)
Adenocarcinoma/therapy , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Nivolumab/therapeutic use , Adenocarcinoma/immunology , Adenocarcinoma/mortality , Adenocarcinoma/pathology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Humans , Immune Checkpoint Inhibitors/adverse effects , Immunotherapy/adverse effects , Neoplasm Staging , Nivolumab/adverse effects , Time Factors , Treatment Outcome
20.
Pathol Oncol Res ; 27: 1609846, 2021.
Article in English | MEDLINE | ID: mdl-34305476

ABSTRACT

A variety of pathogenic microorganisms promote tumor occurrence and development through long-term colonization in the body. Fusobacterium nucleatum (F. nucleatum) is abundant in precancerous esophageal lesions and is closely related to the malignant progression of esophageal squamous cell carcinoma (ESCC). The invasion of exogenous microorganisms can reshape the immune microenvironment, make the immune system incapacitated, and assist tumor cells in immune escape. A variety of pathogenic microorganisms induce the recruitment of regulatory T cell (Tregs) to allow tumor cells to escape immune surveillance and provide favorable conditions for their own long-term colonization. Tregs are one of the major obstacles to tumor immunotherapy and have a significant positive correlation with the occurrence and development of many kinds of tumors. Because F. nucleatum can instantly enter cells and colonize for a long time, we speculated that F. nucleatum infection could facilitate the immune escape of tumor cells through enrichment of Tregs and promote the malignant progression of ESCC. In this study, we found a significant concordance between F. nucleatum infection and Tregs infiltration. Therefore, we propose the view that chronic infection of F. nucleatum may provide favorable conditions for long-term colonization of itself by recruiting Tregs and suppressing the immune response. At the same time, the massive enrichment of Treg may also weaken the immune response and assist in the long-term colonization of F. nucleatum. We analyzed the correlation between F. nucleatum infection with the clinicopathological characteristics and survival prognosis of the patients. F. nucleatum infection was found to be closely related to sex, smoking, drinking, degree of differentiation, depth of invasion, lymph node metastasis, and clinical stage. The degree of differentiation, depth of infiltration, lymph node metastasis, clinical stage, and F. nucleatum infection are independent risk factors affecting ESCC prognosis. Additionally, the survival rate and median survival time were significantly shortened in the F. nucleatum infection positive group. Therefore, we propose that long-term smoking and alcohol consumption cause poor oral and esophageal environments, thereby significantly increasing the risk of F. nucleatum infection. In turn, F. nucleatum infection and colonization may weaken the antitumor immune response through Treg enrichment and further assist in self-colonization, promoting the malignant progression of ESCC.


Subject(s)
Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/pathology , Fusobacterium Infections/complications , Fusobacterium nucleatum/pathogenicity , T-Lymphocytes, Regulatory/immunology , Tumor Microenvironment , Esophageal Neoplasms/immunology , Esophageal Neoplasms/microbiology , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/microbiology , Esophageal Squamous Cell Carcinoma/surgery , Female , Follow-Up Studies , Fusobacterium Infections/microbiology , Humans , Lymphatic Metastasis , Male , Middle Aged , Prognosis , Survival Rate
SELECTION OF CITATIONS
SEARCH DETAIL
...