Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
1.
Bioengineering (Basel) ; 11(4)2024 Apr 18.
Article in English | MEDLINE | ID: mdl-38671811

ABSTRACT

With the further development of neural networks, automatic segmentation techniques for melanoma are becoming increasingly mature, especially under the conditions of abundant hardware resources. This allows for the accuracy of segmentation to be improved by increasing the complexity and computational capacity of the model. However, a new problem arises when it comes to actual applications, as there may not be the high-end hardware available, especially in hospitals and among the general public, who may have limited computing resources. In response to this situation, this paper proposes a lightweight deep learning network that can achieve high segmentation accuracy with minimal resource consumption. We introduce a network called DTONet (double-tailed octave network), which was specifically designed for this purpose. Its computational parameter count is only 30,859, which is 1/256th of the mainstream UNet model. Despite its reduced complexity, DTONet demonstrates superior performance in terms of accuracy, with an IOU improvement over other similar models. To validate the generalization capability of this model, we conducted tests on the PH2 dataset, and the results still outperformed existing models. Therefore, the proposed DTONet network exhibits excellent generalization ability and is sufficiently outstanding.

2.
EClinicalMedicine ; 69: 102471, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38356729

ABSTRACT

Background: Concurrent chemoradiotherapy is the standard nonoperative treatment for locally advanced esophageal squamous cell carcinoma. However, local recurrence is still the main failure pattern, accounting for more than half of all treatment failures, indicating that the sensitivity of radiotherapy still needs to be improved. This trial aimed at demonstrating whether PD-1 inhibitors followed by chemoradiotherapy could promote esophageal tumor vascular normalization, alleviate hypoxia, and thus enhance radiosensitivity and improve local control. Methods: We did a multicenter, single-arm, phase 2 trial in China. Patients with locally advanced esophageal cancer were enrolled in this study. In induction phase, patients received two cycles of sintilimab, paclitaxel and carboplatin once per 21 days. In concurrent phase, patients were treated with five cycles of carboplatin and paclitaxel once per week concurrent with radiotherapy of 50.4Gy delivered in 28 fractions. The primary endpoint was 2-year local control rate. Hypoxia and vessel normalization was assessed before and after induction phase using immunofluorescence and perfusion CT. This trial is registered with ClinicalTrials.gov (NCT03985046). Findings: Seventy-five patients with esophageal cancer were enrolled in this study between October 2019 and April 2021. The median follow-up of surviving patients was 33.6 months (IQR 29.3-35.7). The 2-year local control rate was 81.7% (95% confidence interval, 72.7%-90.7%), which was much higher than that in concurrent chemoradiation only (71.3%) in previous studies. Vascular normalization and hypoxia alleviation were observed in both biopsy specimens and perfusion CT. Interpretation: The addition of induction immunotherapy to standard concurrent chemoradiotherapy could improve radiosensitivity for locally advanced esophageal cancer as non-surgical treatment. New treatment combination led to higher local control rate through promoting vascular normalization and alleviating hypoxia. Our findings suggest that induction immunotherapy followed by concurrent chemoradiotherapy could be a potential option in future treatment. Funding: National Natural Science Foundation of China and Shanghai Rising-Star Program.

3.
Lancet Gastroenterol Hepatol ; 9(1): 45-55, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37980921

ABSTRACT

BACKGROUND: The efficacy of local therapy for patients with oligometastatic oesophageal squamous cell carcinoma is unclear. We aimed to assess the efficacy of local plus systemic therapy compared with systemic therapy alone in patients with oligometastatic oesophageal squamous cell carcinoma. METHODS: The ESO-Shanghai 13 trial was a randomised, open-label, multicentre, phase 2 trial. Patients (aged ≥18 years) were recruited from six hospitals in China with histological confirmation of oligometastatic oesophageal squamous cell carcinoma with a controlled primary tumour and one to four metastatic lesions. Eligible patients were randomly assigned via a computer-generated schedule in a 1:1 ratio to receive either systemic therapy alone (ie, systemic therapy only group) or combined systemic and local therapy (ie, systemic and local therapy group). The systemic therapy regimens in both groups were at the discretion of the investigator and included chemotherapy alone, anti-PD-1 antibodies alone, or chemotherapy plus anti-PD-1 antibodies. Local therapy-radiotherapy, surgery, or thermal ablation-was delivered to all metastatic lesions for patients in the systemic and local therapy group. Randomisation was balanced dynamically on three factors: the number of disease sites, the lines of systemic therapy, and the location of the metastases. Patients and investigators were not masked to treatment allocation. The primary endpoint was progression-free survival, defined as the time from randomisation to progression or death from any cause in the intention-to-treat population. The safety population included all patients who had undergone random assignment and at least one of the intended therapies. This trial is registered with ClinicalTrials.gov, NCT03904927. The trial is ongoing but closed to new participants. FINDINGS: 116 patients were screened for enrolment between March 5, 2019, and Sept 16, 2021, and 104 patients who met the eligibility criteria were randomly assigned to the systemic and local therapy group (n=53) or the systemic therapy only group (n=51). 20 (38%) patients in the systemic plus local therapy group and 23 (45%) patients in the systemic therapy only group received anti-PD-1 antibody-based systemic therapy; three patients in the systemic and local therapy group did not receive systemic therapy. At a median follow-up of 30·5 months (IQR 24·7-37·8), median progression-free survival was 15·3 months (95% CI 10·1-20·5) in the systemic and local therapy group versus 6·4 months (5·2-7·6) in the systemic therapy only group (stratified hazard ratio 0·26 [95% CI 0·16-0·42]; stratified log rank p<0·0001). Grade 1-2 acute oesophagitis was more common in the systemic and local therapy group than in the systemic therapy only group (10 [19%] vs one [2%] patients; p=0·036). The number of patients who had grade 3 or worse treatment-related adverse events was similar between groups (25 [47%] vs 21 [41%]; p=0·538), with the most common adverse events being leukocytopenia (17 [32%] vs 18 [35%]) and neutropenia (19 [36%] vs 20 [39%]). Treatment-related deaths occurred in two patients in the systemic and local therapy group and one patient in the systemic therapy only group. INTERPRETATION: The addition of local treatment for metastases could significantly improve progression-free survival among patients with oligometastatic oesophageal squamous cell carcinoma being treated with systemic therapy. Our findings suggest that combining local and systemic therapy could be a treatment option for patients with oligometastatic oesophageal squamous cell carcinoma, but further support from phase 3 trials is required. FUNDING: Science and Technology Commission of Shanghai Municipality, National Nature Science Foundation of China, and Shanghai Municipal Health Commission. TRANSLATION: For the Chinese translation of the abstract see Supplementary Materials section.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Adolescent , Adult , Esophageal Squamous Cell Carcinoma/drug therapy , China/epidemiology , Progression-Free Survival , Proportional Hazards Models , Esophageal Neoplasms/drug therapy
4.
Radiat Oncol ; 18(1): 172, 2023 Oct 19.
Article in English | MEDLINE | ID: mdl-37858121

ABSTRACT

BACKGROUND: Limited studies explored the relationship between lymphocyte recovery after definitive concurrent chemoradiotherapy (dCCRT) and prognosis in esophageal squamous cell carcinoma (ESCC). METHODS: ESCC patients with obtainable absolute lymphocyte counts (ALCs) at 6 months after dCCRT were screened from prospective trials. Patients were divided into groups according to the grade of ALC nadir during radiotherapy (G4 or G1-3) and lymphocyte recovery status, which was assessed by lymphocyte recovery index (LRI), calculated as the ratio of post- to pre-treatment lymphocyte counts. Cox analysis was conducted to evaluate the prognostic significance of lymphocyte recovery status. Irradiated relative volumes of the bone marrow (BM) and spleen and effective dose to immune cells (EDIC) were collected to identify their impacts on lymphocyte recovery status by logistic analysis. RESULTS: 232 patients were enrolled. In 69 patients with G4 ALC nadir (group A and B) and 163 patients with G1-3 ALC nadir (group C and D) during dCCRT, 27 (group A) and 67 (group C) patients showed an insufficient level of lymphocyte recovery (LRI < 60%), and 42 (group B) and 96 (group D) patients showed a satisfactory level of lymphocyte recovery (LRI ≥ 60%). Cox multivariable analysis revealed that inadequate lymphocyte recovery was significantly associated with worse overall survival (HR, 2.80 and 1.70) and local recurrence-free survival (HR, 2.82 and 1.60) both in group A vs group B and group C vs group D. Logistic analysis identified BM V5 (OR 4.24 and 2.29) as an independent predictor of inadequate lymphocyte recovery from G4 or G1-3 ALC nadir, respectively. CONCLUSIONS: Insufficient lymphocyte recovery might serve as a valuable prognostic factor, regardless of whether patients experienced G4 or G1-3 ALC nadir during radiotherapy. Additionally, it was observed that a larger relative volume of BM receiving ≥ 5 Gy was correlated with a higher risk of insufficient lymphocyte recovery.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lymphopenia , Humans , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/pathology , Prospective Studies , Lymphopenia/pathology , Lymphocytes/pathology , Prognosis , Chemoradiotherapy/adverse effects , Retrospective Studies
5.
Quant Imaging Med Surg ; 13(9): 6280-6295, 2023 Sep 01.
Article in English | MEDLINE | ID: mdl-37711778

ABSTRACT

Background: Among cancers, esophageal cancer (EC) has one of the highest incidences and mortality in Asia. As recognized in many national guidelines, functional imaging performed with position emission tomography is recommended for patients with locally advanced disease. This review evaluated evidence for the use of fluorodeoxyglucose (FDG) interim positron emission tomography (PETint) in bimodality (chemoradiation) and trimodality (chemoradiation followed by surgery) management of locally advanced esophageal cancer (LAEC), with a focus on its prognostic and predictive value. Methods: The MEDLINE database was searched from January 1, 2001, to January 1, 2022, as part of a scoping review. References of selected articles were manually checked to identify other articles meeting the inclusion criteria; only original articles were included, and reviews, guidelines, letters, editorials, and case reports were excluded. Results: A total of 63 articles were included in this review. PET-computed tomography (PET-CT) is recognized as having a significant role in the assessment of treatment response. Studies on the predictive PETint suggest that it has a certain value, particularly for early response. Identification of poor responders or nonresponders soon after commencement of multimodality treatment allows for treatment modification. Conclusions: The scoping review indicated variable utility for the prognostic value of PETint. There is a need to improve its accuracy, which can likely be achieved through greater standardization of measurements and reporting and testing as well as combination with other promising measures of response to residual disease.

6.
Oncologist ; 28(8): e645-e652, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37285035

ABSTRACT

BACKGROUND: This study aimed to explore the relationship between irradiation of lymphocyte-related organs at risk (LOARs) and lymphopenia during definitive concurrent chemoradiotherapy (dCCRT) for esophageal squamous cell carcinoma (ESCC). MATERIALS AND METHODS: Cases of ESCC patients who received dCCRT from 2 prospective clinical trials were identified. To find its correlation with survival outcomes, grades of absolute lymphocyte counts (ALCs) nadir during radiotherapy were recorded following COX analysis. Associations of lymphocytes at nadir and dosimetric parameters including relative volumes of spleen and bone marrow receiving 0.5, 1, 2, 3, 5, 10, 20, 30, and 50Gy (V0.5, V1, V2, V3, V5, V10, V20, V30, and V50), and effective dose to circulating immune cells (EDIC) were examined by logistic risk regression analysis. The cutoffs of dosimetric parameters were determined by the receiver operating characteristic curve (ROC). RESULTS: A total of 556 patients were included. The incidences of grades 0, 1, 2, 3, and 4 (G4) lymphopenia during dCCRT were 0.2%, 0.5%, 9.7%, 59.7%, and 29.8%, respectively. Their median overall survival (OS) and progression-free survival (PFS) time were 50.2 and 24.3 months, respectively; the incidence of local recurrence and distant metastasis were 36.6% and 31.8%, respectively. Patients once suffering from G4 nadir during radiotherapy had unfavorable OS (HR, 1.28; P = .044) and a higher incidence of distant metastasis (HR, 1.52; P = .013). Furthermore, patients with EDIC ≤8.3Gy plus spleen V0.5 ≤11.1% and bone marrow V10 ≤33.2% were strongly associated with lower risk of G4 nadir (OR, 0.41; P = .004), better OS (HR, 0.71; P = .011) and lower risk of distant metastasis (HR, 0.56; P = .002). CONCLUSIONS: Smaller relative volumes of spleen V0.5 and bone marrow V10 plus lower EDIC were jointly prone to reduce the incidence of G4 nadir during definitive concurrent chemoradiotherapy. This modified therapeutic strategy could be a significant prognostic factor for survival outcomes in ESCC.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Lymphopenia , Humans , Esophageal Neoplasms/complications , Esophageal Neoplasms/radiotherapy , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/radiotherapy , Esophageal Squamous Cell Carcinoma/pathology , Prospective Studies , Lymphopenia/etiology , Lymphopenia/pathology , Chemoradiotherapy/adverse effects , Lymphocytes/pathology , Retrospective Studies
7.
Radiat Res ; 198(5): 458-466, 2022 11 01.
Article in English | MEDLINE | ID: mdl-35993821

ABSTRACT

Radiotherapy is a main treatment for esophageal squamous cell carcinoma (ESCC), but radioresistance leads to treatment failure ultimately. The combination of radiotherapy and PD-1 inhibitors showed significant antitumor effects. Our study showed that high-immune score, IFNG and CD8A level were associated with a low-radiosensitivity index (RSI) in the TCGA-ESCC cohort. And blocking PD-1 promoted exhausted T cells proliferation and IFN-γ expression. PD-1 inhibitor-reactivated T cells promoted G2/M-phase arrest, apoptosis and impaired DNA damage in radioresistant cells in an IFN-γ-dependent manner. Our study showed PD-1 inhibitors promote radiosensitivity though enhancing exhausted T cells expansion and IFN-γ expression, and highlights that neoadjuvant anti-PD-1 therapy and radiotherapy could offer an optimum strategy for improving cancer patients' outcome.


Subject(s)
Carcinoma, Squamous Cell , Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Humans , Esophageal Squamous Cell Carcinoma/radiotherapy , Immune Checkpoint Inhibitors , Esophageal Neoplasms/drug therapy , Carcinoma, Squamous Cell/radiotherapy , Carcinoma, Squamous Cell/drug therapy , Cell Line, Tumor , T-Lymphocytes/metabolism , Radiation Tolerance , Cell Proliferation , Apoptosis , Mitosis
8.
Radiat Oncol ; 17(1): 134, 2022 Jul 29.
Article in English | MEDLINE | ID: mdl-35906623

ABSTRACT

INTRODUCTION: Definitive chemoradiotherapy has established the standard non-surgical treatment for locally advanced esophageal cancer. The standard dose of 50-50.4 Gy has been established decades ago and been confirmed in modern trials. The theorical advantage of better local control and technical advances for less toxicity have encouraged clinicians for dose escalation investigation. 18F-fluorodeoxyglucose (18F-FDG) positron emission tomography/computed tomography (PET/CT) have the potential to tailor therapy for esophageal patients not showing response to CRT and pioneers the PET-based dose escalation. METHODS AND ANALYSIS: The ESO-Shanghai 12 trial is a prospective multicenter randomized phase 3 study in which patients are randomized to either 61.2 Gy or 50.4 Gy of radiation dose by PET response. Both groups undergo concurrent chemoradiotherapy with paclitaxel/cisplatin regimen for 2 cycles followed by consolidation chemotherapy for 2 cycles. Patients with histologically confirmed ESCC [T1N1-3M0, T2-4NxM0, TxNxM1 (Supraclavicular lymph node metastasis only), (AJCC Cancer Staging Manual, 8th Edition)] and without any prior treatment of chemotherapy, radiotherapy or surgery against esophageal cancer will be eligible. The primary endpoints included overall survival in PET/CT non-responders (SUVmax > 4.0) and overall survival in total population. Patients will be stratified by standardized uptake volume, gross tumor volume and tumor location. The enrollment could be ended, when the number of PET/CT non-responder reached 132 and the total population reached 646 for randomization. ETHICS AND DISSEMINATION: This trial has been approved by the Fudan University Shanghai Cancer Center Institutional Review Board. Trial results will be disseminated via peer reviewed scientific journals and conference presentations. Trial registration The trial was initiated in 2018 and is currently recruiting patients. Trial registration number NCT03790553.


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Chemoradiotherapy , China , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/therapy , Esophageal Squamous Cell Carcinoma/therapy , Fluorodeoxyglucose F18 , Humans , Positron Emission Tomography Computed Tomography/methods , Prospective Studies , Radiopharmaceuticals , Treatment Outcome
9.
Curr Med Chem ; 27(30): 4932-4944, 2020.
Article in English | MEDLINE | ID: mdl-30827229

ABSTRACT

Different natural and synthetic biodegradable polymers have been used in vaccine formulations as adjuvant and delivery system but have faced various limitations. Chitosan is a new delivery system with the potential to improve development of nano vaccines and drugs. However, chitosan is only soluble in acidic solutions of low concentration inorganic acids such as dilute acetic acid and dilute hydrochloric acid and in pure organic solvents, which greatly limits its application. Chemical modification of chitosan is an important way to improve its weak solubility. Quaternized chitosan not only retains the excellent properties of chitosan, but also improves its water solubility for a wider application. Recently, quaternized chitosan nanoparticles have been widely used in biomedical field. This review focuses on some quaternized chitosan nanoparticles, and points out the advantages and research direction of quaternized chitosan nanoparticles. As shown by the applications of quaternized chitosan nanoparticles as adjuvant and delivery carrier in vaccines, quaternized chitosan nanoparticles have promising potential in application for the development of nano vaccines in the future.


Subject(s)
Chitosan , Nanoparticles , Vaccines , Adjuvants, Immunologic , Polymers
10.
Nat Commun ; 10(1): 4601, 2019 10 10.
Article in English | MEDLINE | ID: mdl-31601798

ABSTRACT

During pregnancy, trophoblast cells sustain the maternal-fetal tolerance via expressing and secreting various chemokines and cytokines. Our previous study revealed the expression of interleukin-35 (IL-35) in human first-trimester trophoblasts. Here we show that IL-35 is expressed in both human first-trimester primary trophoblast cells and a trophoblast cell line. Trophoblast cells inhibit the proliferation of human naive conventional T cells (Tconv cells) and convert suppressed Tconv cells into iTR35 in an IL-35-dependent manner. Mechanistically, trophoblast cell derived IL-35 mediates its function through phosphorylation of STAT1 and STAT3. In vivo studies confirm that mice with immunologically spontaneous abortion have lower levels of IL-35 and iTR35 cells at the maternal-fetal interface, and neutralizing anti-IL-35 mAb enhances abortion rates. Meanwhile, exogenous IL-35 induces iTR35 and prevents immunological abortion. Our findings thus suggest that trophoblast cells have a critical function in preserving maternal-fetal tolerance via secreting IL-35 during pregnancy.


Subject(s)
Interleukins/metabolism , Maternal-Fetal Exchange/physiology , T-Lymphocytes, Regulatory/immunology , Trophoblasts/cytology , Animals , Cell Proliferation , Female , Humans , Immune Tolerance , Interleukins/blood , Interleukins/immunology , Interleukins/pharmacology , Male , Maternal-Fetal Exchange/immunology , Mice, Inbred BALB C , Mice, Inbred DBA , Placenta/cytology , Placenta/immunology , Pregnancy , Recombinant Proteins/genetics , Recombinant Proteins/immunology , Recombinant Proteins/pharmacology , T-Lymphocytes/cytology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism , Trophoblasts/immunology , Trophoblasts/metabolism
11.
Exp Cell Res ; 383(2): 111513, 2019 10 15.
Article in English | MEDLINE | ID: mdl-31362000

ABSTRACT

Regulatory B cells (Breg cells) play critical roles in modulating immune responses during autoimmune diseases and infection. Here we explored the participation of two main Breg subsets, including IL-10+ Breg (B10) and IL-35+ Breg cells, in maintaining successful pregnancy. We first observed an elevated percentage of B10 cells in peripheral blood from first-trimester pregnant women compared with non-pregnant controls. Serum from pregnancy induced the augmentation of B10  in peripheral blood mononuclear cells from non-pregnant women. In animal models, we demonstrated that there were significant augmentation of B10 cells and obvious increase of IL-10 level in splenic B cells from normal pregnant mice compared to that in abortion-prone pregnant mice and virgin mice. Further analysis showed that both hCG and IL-35 suppressed the proliferation of mouse splenic B cells. Moreover, IL-35 induced the expansion of both mouse splenic B10 and IL-35+ Breg cells while hCG only mediated the generation of B10 cells. Subsequent study in mice demonstrated that the activation of STAT1 and STAT3 in B cells caused by IL-35 and the activation of STAT3 caused by hCG were the predominant mechanism of IL-35+ Breg and B10 cells augmentation. These findings suggested that hCG and IL-35 induced the amplification of B10 and IL-35+ Breg cells which played a vital peripheral regulatory role during pregnancy.


Subject(s)
B-Lymphocytes, Regulatory/physiology , Chorionic Gonadotropin/physiology , Immune Tolerance , Interleukin-10/metabolism , Interleukins/physiology , Pregnancy Maintenance/immunology , Abortion, Spontaneous/blood , Abortion, Spontaneous/immunology , Abortion, Spontaneous/pathology , Adult , Animals , B-Lymphocytes, Regulatory/drug effects , B-Lymphocytes, Regulatory/metabolism , Case-Control Studies , Cell Proliferation/drug effects , Cells, Cultured , Chorionic Gonadotropin/blood , Chorionic Gonadotropin/pharmacology , Disease Models, Animal , Female , Humans , Immune Tolerance/drug effects , Interleukins/blood , Interleukins/pharmacology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred CBA , Mice, Inbred DBA , Pregnancy , Pregnancy Maintenance/drug effects , Pregnancy Trimester, First/blood , Pregnancy Trimester, First/immunology , Young Adult
12.
Carcinogenesis ; 39(12): 1488-1496, 2018 12 31.
Article in English | MEDLINE | ID: mdl-30321288

ABSTRACT

Interleukin 35 (IL-35) is a potent immunosuppressive cytokine, consisting of an Epstein-Barr virus-induced gene 3 (EBI3) subunit and a p35 subunit. IL-35 is mainly produced by regulatory T and regulatory B cells, and plays a crucial role in the development and prevention of infectious and autoimmune diseases. However, the effect of IL-35 in malignant disease is not well understood. In this study, we demonstrated that breast cancer cells (BCCs) also expressed and secreted IL-35 and higher level of IL-35 in BCCs was closely associated with poor prognosis of patients and was an independent unfavorable prognostic factor for breast cancer. Subsequent study revealed that BCC-derived IL-35 inhibited conventional T (Tconv) cell proliferation and further induced suppressed Tconv cells into IL-35-producing induced regulatory T (iTr35) cells. Furthermore, BCC-derived IL-35 promoted the secretion of inhibitory cytokine IL-10 and obviously decreased the secretion of Th1-type cytokine IFN-γ and Th17-type cytokine IL-17 in Tconv cells. Meanwhile, the expression of inhibitory receptor CD73 was also elevated on the surface of Tconv cells following the BCCs' supernatant treatment. Mechanistically, BCC-derived IL-35 exhausted Tconv cells and induced iTr35 by activating transcription factor STAT1/STAT3. Hence, our results indicate functions of BCC-derived IL-35 in promoting tumor progression through proliferation inhibition of tumor-infiltrating Tconv cells and induction of iTr35 cells in tumor microenvironment. This study highlights that IL-35 produced by BCCs are a potential therapeutic target for breast cancer.


Subject(s)
Breast Neoplasms/immunology , Interleukins/immunology , T-Lymphocytes, Regulatory/immunology , Breast Neoplasms/pathology , Cell Line , Cell Line, Tumor , Cell Proliferation/physiology , Disease Progression , Female , HEK293 Cells , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Lymphocyte Activation/immunology , MCF-7 Cells , Middle Aged , STAT1 Transcription Factor/immunology , STAT3 Transcription Factor/immunology , T-Lymphocytes, Regulatory/pathology , Tumor Microenvironment/immunology
14.
Cytokine ; 108: 43-52, 2018 08.
Article in English | MEDLINE | ID: mdl-29571039

ABSTRACT

IL-35, a novel IL-12 family member, is a potent inhibitory cytokine predominantly produced by regulatory T and B lymphocytes that exerts optimal suppression in immune response. However, it remains unclear whether IL-35 plays an inhibitory role on human dendritic cells. In the present study, we focused on the possible immunosuppressive effect of IL-35 on the differentiation, maturation and function of monocyte-derived DCs (MoDCs). Addition of exogenous IL-35 was able to partially suppress MoDCs differentiation in vitro. Subsequently, LPS was used for the maturation of MoDCs and IL-35 was found to mainly restrain the maturation of MoDCs, characterized by the remarkable down-regulation of costimulatory molecules, CD83 and HLA-DR as well as a reduced production of pro-inflammatory cytokines (IL-12p70, IFN-γ, and TNF-α). Furthermore, IL-35-treated MoDCs exhibited strong inhibition in the proliferation of allogeneic CD4+/CD8+ T lymphocytes. Meanwhile, IL-35-treated MoDCs also suppressed the polarization of naïve CD4+ T lymphocytes towards Th1 phenotype and impaired CD8+ T cells allogeneic responses. And the foregoing suppression of MoDCs maturation and function by IL-35 might be due to the aberrant activation of STAT1/STAT3 and inhibition of p38 MAPK/NF-κB signaling pathway. Our results demonstrated for the first time that IL-35 played a critical role in modulating not only adaptive immune response, but also innate immune response. The inhibitory effect of IL-35 on MoDCs maturation and function may facilitate the development of promising therapeutic interventions in tumors and other diseases.


Subject(s)
Cell Differentiation/drug effects , Dendritic Cells/physiology , Interleukins/genetics , Interleukins/immunology , Lymphocyte Activation/drug effects , Adaptive Immunity/drug effects , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes/immunology , Cell Differentiation/immunology , Cells, Cultured , Coculture Techniques , Cytokines/analysis , Cytokines/immunology , Dendritic Cells/drug effects , Humans , Immunity, Innate/drug effects , Interleukins/pharmacology , Lipopolysaccharides/pharmacology , Lymphocyte Activation/immunology , Monocytes/immunology , Monocytes/physiology , Th1 Cells/immunology , Th1 Cells/physiology
15.
Oncotarget ; 8(8): 13703-13715, 2017 Feb 21.
Article in English | MEDLINE | ID: mdl-28099147

ABSTRACT

Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4), a potent immunoregulatory molecule, can down-regulate T-cell activation and inhibit anti-tumor immune response. This study showed that LPS-stimulated human dendritic cells (DCs) decreased the expression of HLA-DR, CD83 and costimulatory molecules (CD40, CD80 and CD86) following coculturing with CTLA-4+ breast cancer cells. Moreover, the suppressed DCs further inhibited proliferation of allogeneic CD4+/CD8+ T-cells, differentiation of Th1 and function of cytotoxic lymphocytes (CTLs). However, CTLA-4 blockade in breast cancer cells could recover DC maturation and cytokine production, elevate antigen-presenting function of DCs, reverse Th1/CTLs response and cytokine secretion. Subsequent study demonstrated that the activation of extracellular-signal regulated kinase and signal transducer and activator of transcription 3 of DCs caused by CTLA-4+ breast cancer cells were the predominant mechanism of DC suppression. In addition, CTLA-4 blockade treatment also directly inhibited proliferation and induced apoptosis of CTLA-4+ breast cancer cells. Collectively, CTLA-4 was expressed and functional on human breast cancer cells through influencing maturation and function of DCs in vitro, and CTLA-4 blockage not only recovered the antigen-presenting function of DCs and T-cells activation but also suppressed the biological activity of breast cancer cells themselves. This study highlights the clinical application of CTLA-4 blockade therapy in breast cancer.


Subject(s)
Breast Neoplasms/immunology , CTLA-4 Antigen/immunology , Dendritic Cells/immunology , Antigen Presentation , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/pathology , CTLA-4 Antigen/antagonists & inhibitors , CTLA-4 Antigen/biosynthesis , Cytokines/biosynthesis , Cytokines/immunology , Dendritic Cells/pathology , Female , Flow Cytometry , Humans , MCF-7 Cells , Monocytes/immunology , Monocytes/pathology , Th1 Cells/immunology , Th1 Cells/pathology
16.
Cytokine ; 89: 76-81, 2017 01.
Article in English | MEDLINE | ID: mdl-27681506

ABSTRACT

Interleukin-35 (IL-35) is a recently discovered inhibitory cytokine, which is firstly discovered to be produced by regulatory T cells (Tregs) and proposed as a key effector molecule of Treg function. This study aims to analyze the correlation between IL-35 expression in tumor-infiltrating lymphocytes (TILs) of breast cancer tissue and patients' clinical characteristics. Plasma IL-35 was also determined in 60 patients with breast invasive ductal carcinoma (IDC) and 30 healthy women by enzyme-linked immunosorbent assay. IL-35 expression in the tissue specimens was analyzed by immunohistochemistry. It was shown that 39.1%, 43.6% and 17.3% of the 110 patients were absent, weak, and strong IL-35 expression in the TILs, respectively. Strong IL-35 expression in TILs was significantly associated with age >50years, tumor size >2cm, TNM stage III, and negative ER (All P<0.05). Patients with elevated IL-35 expression in TILs had significantly worse progression-free survival (PFS) and overall survival (OS) than patients with weak or no IL-35 expression (All P<0.05). High plasma IL-35 levels were significantly associated with TNM stage III and lymph node metastasis (All P<0.05). Plasma IL-35 level and IL-35 expression in the TILs of breast cancer tissues may be a valuable biomarker in the development and prognosis of IDC.


Subject(s)
Biomarkers, Tumor/biosynthesis , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Gene Expression Regulation, Neoplastic , Interleukins/biosynthesis , Neoplasm Proteins/biosynthesis , T-Lymphocytes, Regulatory/metabolism , Adult , Breast Neoplasms/pathology , Breast Neoplasms/therapy , Disease-Free Survival , Female , Humans , Middle Aged , Survival Rate , T-Lymphocytes, Regulatory/pathology
17.
Cancer Lett ; 373(1): 97-108, 2016 04 01.
Article in English | MEDLINE | ID: mdl-26806810

ABSTRACT

This article has been retracted: please see Elsevier Policy on Article Withdrawal (http://www.elsevier.com/locate/withdrawalpolicy). This article has been retracted at the request of the Editor in Chief. The term miR-143 has been used instead of the term miR-143* in the introduction and discussion which is misleading. With the misuse of this term subsequent errors and misleading statements appear throughout the paper. The authors apologize for this mistake.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Cell Movement , Cell Proliferation , Esophageal Neoplasms/metabolism , MicroRNAs/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction , 3' Untranslated Regions , Animals , Binding Sites , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/mortality , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Epithelial-Mesenchymal Transition , Esophageal Neoplasms/genetics , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma , Female , G1 Phase Cell Cycle Checkpoints , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Male , Mice, Inbred BALB C , Mice, Nude , MicroRNAs/genetics , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , STAT3 Transcription Factor/genetics , Time Factors , Transfection
18.
J Magn Reson Imaging ; 44(2): 445-55, 2016 08.
Article in English | MEDLINE | ID: mdl-26778191

ABSTRACT

PURPOSE: To explore the clinical potential of texture analysis using contrast-enhanced 3.0T magnetic resonance imaging (MRI) for predicting the therapeutic response of nasopharyngeal carcinoma (NPC) to chemoradiotherapy. MATERIALS AND METHODS: The dataset comprised pretreatment T1 -, T2 -, and diffusion-weighted MR images from 53 eligible patients with newly diagnosed NPC. The patients were divided into two sets: the training set including 31 responders and 11 nonresponders and the testing set including eight responders and three nonresponders. The region of interest (ROI) was delineated by two radiologists for each sequence. Quantitative image parameters were extracted and statistically filtered to identify a subset of reproducible and nonredundant parameters that were used to construct the predictive model. The internal validation was performed using stratified 10-fold cross-validation in the training set and the external validation was performed in the testing set. McNemar's test was used to test the statistical difference between the performances of the extracted parameters in predicting the treatment response. RESULTS: All three parameter sets showed potential in predicting treatment response with high accuracy (T1 : 0.952/0.939, T2 : 0.904/0.905, diffusion-weighted [DWI]: 0.881/0.929). Supervised learning models based on parameters extracted from the T1 sequence showed better classification performance than those extracted from the T2 -weighted (T2 W) (artificial neural network [ANN]: P = 0.043, k-nearest neighbors [kNN]: P = 0.033) and DWI (ANN: P = 0.032. kNN: P = 0.014). No statistical difference was observed in the performance of the two classifiers (P = 0.083). CONCLUSION: Texture analysis based on T1 W, T2 W, and DWI could act as imaging biomarkers of tumor response to chemoradiotherapy in NPC patients and serve as a new radiological analysis tool for treatment prediction. J. Magn. Reson. Imaging 2016;44:445-455.


Subject(s)
Carcinoma/drug therapy , Carcinoma/therapy , Chemoradiotherapy/methods , Image Interpretation, Computer-Assisted/methods , Imaging, Three-Dimensional/methods , Magnetic Resonance Imaging/methods , Nasopharyngeal Neoplasms/drug therapy , Nasopharyngeal Neoplasms/therapy , Outcome Assessment, Health Care/methods , Adult , Aged , Carcinoma/pathology , Contrast Media , Female , Humans , Image Enhancement/methods , Male , Middle Aged , Nasopharyngeal Carcinoma , Nasopharyngeal Neoplasms/pathology , Reproducibility of Results , Sensitivity and Specificity , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...