Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 94
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Nat Immunol ; 22(3): 358-369, 2021 03.
Article in English | MEDLINE | ID: mdl-33432230

ABSTRACT

CD8+ T cell exhaustion dampens antitumor immunity. Although several transcription factors have been identified that regulate T cell exhaustion, the molecular mechanisms by which CD8+ T cells are triggered to enter an exhausted state remain unclear. Here, we show that interleukin-2 (IL-2) acts as an environmental cue to induce CD8+ T cell exhaustion within tumor microenvironments. We find that a continuously high level of IL-2 leads to the persistent activation of STAT5 in CD8+ T cells, which in turn induces strong expression of tryptophan hydroxylase 1, thus catalyzing the conversion to tryptophan to 5-hydroxytryptophan (5-HTP). 5-HTP subsequently activates AhR nuclear translocation, causing a coordinated upregulation of inhibitory receptors and downregulation of cytokine and effector-molecule production, thereby rendering T cells dysfunctional in the tumor microenvironment. This molecular pathway is not only present in mouse tumor models but is also observed in people with cancer, identifying IL-2 as a novel inducer of T cell exhaustion.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/metabolism , CD8-Positive T-Lymphocytes/drug effects , Interleukin-2/metabolism , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/metabolism , Receptors, Aryl Hydrocarbon/metabolism , Tumor Microenvironment , 5-Hydroxytryptophan/metabolism , Animals , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Basic Helix-Loop-Helix Transcription Factors/deficiency , Basic Helix-Loop-Helix Transcription Factors/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Gene Expression Regulation, Neoplastic , HCT116 Cells , HEK293 Cells , Humans , Interleukin-2/antagonists & inhibitors , Interleukin-2/genetics , Jurkat Cells , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , MCF-7 Cells , Melanoma, Experimental/drug therapy , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/pathology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NIH 3T3 Cells , Neoplasms/drug therapy , Neoplasms/immunology , Neoplasms/pathology , Receptors, Aryl Hydrocarbon/deficiency , Receptors, Aryl Hydrocarbon/genetics , Signal Transduction , Tryptophan Hydroxylase/metabolism , Xenograft Model Antitumor Assays
2.
Sensors (Basel) ; 24(11)2024 Jun 03.
Article in English | MEDLINE | ID: mdl-38894399

ABSTRACT

Defect detection is an indispensable part of the industrial intelligence process. The introduction of the DETR model marked the successful application of a transformer for defect detection, achieving true end-to-end detection. However, due to the complexity of defective backgrounds, low resolutions can lead to a lack of image detail control and slow convergence of the DETR model. To address these issues, we proposed a defect detection method based on an improved DETR model, called the GM-DETR. We optimized the DETR model by integrating GAM global attention with CNN feature extraction and matching features. This optimization process reduces the defect information diffusion and enhances the global feature interaction, improving the neural network's performance and ability to recognize target defects in complex backgrounds. Next, to filter out unnecessary model parameters, we proposed a layer pruning strategy to optimize the decoding layer, thereby reducing the model's parameter count. In addition, to address the issue of poor sensitivity of the original loss function to small differences in defect targets, we replaced the L1 loss in the original loss function with MSE loss to accelerate the network's convergence speed and improve the model's recognition accuracy. We conducted experiments on a dataset of road pothole defects to further validate the effectiveness of the GM-DETR model. The results demonstrate that the improved model exhibits better performance, with an increase in average precision of 4.9% (mAP@0.5), while reducing the parameter count by 12.9%.

3.
Cancer Immunol Immunother ; 72(1): 23-37, 2023 Jan.
Article in English | MEDLINE | ID: mdl-35661905

ABSTRACT

PURPOSE: Immune checkpoint inhibitors (ICIs) have shown durable responses in various malignancies. However, the response to ICI therapy is unpredictable, and investigation of predictive biomarkers needs to be improved. EXPERIMENTAL DESIGN: In total, 120 patients receiving ICI therapy and 40 patients receiving non-ICI therapy were enrolled. Peripheral blood immune cell markers (PBIMs), as liquid biopsy biomarkers, were analyzed by flow cytometry before ICI therapy, and before the first evaluation. In the ICI cohort, patients were randomly divided into training (n = 91) and validation (n = 29) cohorts. Machine learning algorithms were applied to construct the prognostic and predictive immune-related models. RESULTS: Using the training cohort, a peripheral blood immune cell-based signature (BICS) based on four hub PBIMs was developed. In both the training and the validation cohorts, and the whole cohort, the BICS achieved a high accuracy for predicting overall survival (OS) benefit. The high-BICS group had significantly shorter progression-free survival and OS than the low-BICS group. The BICS demonstrated the predictive ability of patients to achieve durable clinical outcomes. By integrating these PBIMs, we further constructed and validated the support vector machine-recursive and feature elimination classifier model, which robustly predicts patients who will achieve optimal clinical benefit. CONCLUSIONS: Dynamic PBIM-based monitoring as a noninvasive, cost-effective, highly specific and sensitive biomarker has broad potential for prognostic and predictive utility in patients receiving ICI therapy.


Subject(s)
Immune Checkpoint Inhibitors , Neoplasms , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Neoplasms/drug therapy , Algorithms , Flow Cytometry , Liquid Biopsy
4.
Cancer Immunol Immunother ; 72(2): 437-448, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35931835

ABSTRACT

BACKGROUND: Anti-angiogenic drugs increase anti-tumor efficacy of immune checkpoint inhibitors (ICIs). However, the optimal dose of anti-angiogenic drugs remains unclear. METHODS: We retrospectively analyzed efficacy and safety data from patients diagnosed with advanced or metastatic non-small cell lung cancer (NSCLC) that received PD-1 blockade with low-doses of anlotinib, a highly selective receptor tyrosine kinase inhibitor mainly targeting vascular endothelial growth factor receptors, as second or later line therapy. The primary endpoint was progression-free survival (PFS). Secondary endpoints included overall survival (OS), overall response rate (ORR), disease control rate (DCR), and safety profile. Univariate and multivariate analyses were used to identify prognostic factors. RESULTS: A total of 40 eligible patients were included. The median PFS was 11.4 months. The median OS of the entire cohort was 27.0 months. ORR was achieved in 16 patients (40.0%) and DCR was maintained in 33 patients (82.5%). The overall incidence of adverse events (AEs) was 52.5%, and the most common all grade AE was gastrointestinal reactions, which occurred in four patients (10.0%). Treatment-related grade 3/4 toxicity was observed in one patient (2.5%). Conclusions Low-dose anlotinib may be an effective and well-tolerated anti-angiogenesis partner for combination therapy with ICIs in second-line and later settings for advanced NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Quinolines , Humans , Angiogenesis Inhibitors/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/drug therapy , Retrospective Studies , Quinolines/therapeutic use , Indoles/therapeutic use
5.
Cancer Immunol Immunother ; 72(2): 385-395, 2023 Feb.
Article in English | MEDLINE | ID: mdl-35907016

ABSTRACT

BACKGROUND: High hepatitis B virus (HBV) DNA level is an independent risk factor for postoperative HBV-associated liver cancer recurrence. We sought to examine whether HBV DNA level and antiviral therapy are associated with survival outcomes in patients with advanced hepatocellular carcinoma (HCC) treated with anti-programmed cell death protein 1 (PD-1)based immunotherapy. METHODS: This single-institution retrospective analysis included 217 patients with advanced HBV-related HCC treated from 1 June 2018, through 30 December 2020. Baseline information was compared between patients with low and high HBV DNA levels. Overall survival (OS) and progression-free survival (PFS) were compared, and univariate and multivariate analyses were applied to identify potential risk factors for oncologic outcomes. RESULTS: The 217 patients included in the analysis had a median survival time of 20.6 months. Of these HBV-associated HCC patients, 165 had known baseline HBV DNA levels. Baseline HBV DNA level was not significantly associated with OS (P = 0.59) or PFS (P = 0.098). Compared to patients who did not receive antiviral therapy, patients who received antiviral therapy had significantly better OS (20.6 vs 11.1 months, P = 0.020), regardless of HBV DNA levels. Moreover, antiviral status (adjusted HR = 0.24, 95% CI 0.094-0.63, P = 0.004) was an independent protective factor for OS in a multivariate analysis of patients with HBV-related HCC. CONCLUSIONS: HBV viral load does not compromise the clinical outcome of patients with HBV-related HCC treated with anti-PD-1-based immunotherapy. The use of antiviral therapy significantly improves survival time of HBV-related HCC patients.


Subject(s)
Carcinoma, Hepatocellular , Hepatitis B, Chronic , Immune Checkpoint Inhibitors , Liver Neoplasms , Humans , Antiviral Agents/therapeutic use , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/virology , DNA, Viral , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/drug therapy , Immune Checkpoint Inhibitors/therapeutic use , Liver Neoplasms/drug therapy , Liver Neoplasms/virology , Neoplasm Recurrence, Local/drug therapy , Retrospective Studies
6.
Immun Ageing ; 20(1): 75, 2023 Dec 15.
Article in English | MEDLINE | ID: mdl-38102684

ABSTRACT

BACKGROUND: Although the concept of declined immune function associated with cancer has been accepted extensively, real-world clinical studies focusing on analysis of the peripheral blood immune changes underlying ageing, immunity and cancer are scarce. METHODS: In this case-control study, we retrospectively analysed 1375 cancer patients and enrolled 275 age and gender matched healthy individuals. Flow cytometry was conducted to assess the immune changes. Further analysis was examined by SPSS 17.0 and GraphPad Prism 9 software. RESULTS: Cancer patients showed obviously decreased CD3+ T, CD3+CD4+ Th, CD3+CD8+ CTL, CD19+ B, CD16+CD56+ NK cell counts and lower percentage of PD-1 (programmed cell death protein-1, PD-1) positive cells than healthy control (P < 0.0001). For cancer patients, the reference range of circulating percentage of PD-1+CD45+ cells, PD-1+CD3+ T cells, PD-1+CD3+CD4+ Th cells and PD-1+CD3+CD8+ CTL (Cytotoxic T Lymphocyte, CTL) were 11.2% (95% CI 10.8%-11.6%), 15.5% (95% CI 14.7%-16.0%), 15.4% (95% CI 14.9%-16.0%) and 14.5% (95% CI 14.0%-15.5%), respectively. Moreover, the reduction of CD3+ T, CD3+CD4+ Th, CD3+CD8+ CTL, CD19+ B cell counts accompanied with age and stage advancing (P < 0.05). CD16+CD56+ NK cells decreased with stage, but elevated in aged and male cancer patients (P < 0.05). Additionally, the percentage of PD-1 positive cells varied across cancer types, raised with age and stage. Head and neck, pancreatic, gynaecological and lung demonstrated a higher level of the percentage of PD-1 positive cells than melanoma, prostate, and breast cancer (P < 0.05). CONCLUSIONS: This study provides the reference range of the percentage of PD-1 positive cells on peripheral blood, confirms the decreased immune cells and a series of immune changes accompanying with cancer, expands our real world evidence to better understand the interactions of ageing, cancer and immunity. Moreover, the circulating percentage of PD-1 positive cells shows similar tumor type distribution with tumor mutational burden (TMB), supports that it maybe a potential predictive biomarker for immune checkpoint inhibitor therapy.

7.
J Oncol Pharm Pract ; 29(4): 986-991, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36120899

ABSTRACT

INTRODUCTION: Immune-related pneumonitis is an uncommon but potentially life-threatening adverse event associated with anti-programmed cell death protein-1 therapy, and has a higher recurrence rate than that of other pneumonitis. Glucocorticoids are the first treatment of choice for patients with immune-related pneumonitis over grade 1. Given the toxicity associated with glucocorticoids, they should be withdrawn gradually as soon as pneumonitis is controlled. However, low-dose glucocorticoids are maintained in some patients to prevent immune-related pneumonitis. CASE REPORT: We report a rare case of a patient with Hodgkin lymphoma who developed grade 2 immune-related pneumonitis, requiring long-term low-dose glucocorticoid maintenance therapy, during which pneumonitis disappeared, and complete response was achieved. MANAGEMENT AND OUTCOME: Tislelizumab treatment was stopped tentatively, and the patient was given prednisone at an initiating dose of 1 mg/kg/d. The cough symptoms were relieved significantly, and pneumonitis was reduced. The prednisone gradually dwindled, but the immune-related pneumonitis was recurrent, requiring prednisone 10 mg daily maintenance therapy. Subsequently, prednisone and tislelizumab were administered simultaneously, and at present, pneumonitis disappeared and the lesions are in complete remission. DISCUSSION: Low-dose glucocorticoids might play an important role in controlling the recurrence and development of immune-related pneumonitis. The dose and course of glucocorticoid in immune-related pneumonitis patients should be individualized to minimize the toxicity of glucocorticoid.


Subject(s)
Hodgkin Disease , Pneumonia , Humans , Prednisone , Glucocorticoids , Pneumonia/chemically induced , Pneumonia/drug therapy , Hodgkin Disease/drug therapy , Remission Induction
8.
Oncologist ; 27(6): e463-e470, 2022 06 08.
Article in English | MEDLINE | ID: mdl-35348754

ABSTRACT

BACKGROUND: The prognosis of patients with metastatic malignant melanoma is very poor and partly due to resistance to conventional chemotherapies. The study's objectives were to assess the activity and tolerability of apatinib, an oral small molecule anti-angiogenesis inhibitor, in patients with recurrent advanced melanoma. METHODS: This was a single-arm, single-center phase II trial. The primary endpoint was progression-free survival (PFS) and the secondary endpoints were objective response rate (ORR), disease control rate (DCR), and overall survival (OS). Eligible patients had received at least one first-line therapy for advanced melanoma and experienced recurrence. Apatinib (500 mg) was orally administered daily. RESULTS: Fifteen patients (V660E BRAF status: 2 mutation, 2 unknown, 11 wild type) were included in the analysis. The median PFS was 4.0 months. There were two major objective responses, for a 13.3% response rate. Eleven patients had stable disease, with a DCR of 86.7%. The median OS was 12.0 months. The most common treatment-related adverse events of any grade were hypertension (80.0%), mucositis oral (33.3%), hand-foot skin reaction (26.7%), and liver function abnormalities, hemorrhage, diarrhea (each 20%). The only grade ≥3 treatment-related adverse effects that occurred in 2 patients was hypertension (6.7%) and mucositis (6.7%). No treatment-related deaths occurred. CONCLUSION: Apatinib showed antitumor activity as a second- or above-line therapy in patients with malignant melanoma. The toxicity was manageable. CLINICALTRIALS.GOV IDENTIFIER: NCT03383237.


Subject(s)
Melanoma , Neoplasm Recurrence, Local , Pyridines , Antineoplastic Agents/therapeutic use , Humans , Hypertension/chemically induced , Melanoma/drug therapy , Mucositis/chemically induced , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/pathology , Pyridines/adverse effects
9.
Cancer Immunol Immunother ; 71(8): 1897-1908, 2022 Aug.
Article in English | MEDLINE | ID: mdl-34984540

ABSTRACT

BACKGROUND: This first-in-human phase 1 trial is to evaluate the safety, pharmacokinetics, preliminary efficacy, and biomarkers of sugemalimab, a full-length, fully human anti-PD-L1 monoclonal antibody, in Chinese patients with advanced malignancies. METHODS: Eligible patients with unresectable advanced or metastatic solid tumors or lymphomas were enrolled in phase 1a to receive sugemalimab following a modified 3 + 3 design. The primary endpoints included safety, tolerability, and the recommended Phase 2 dose (RP2D). In phase 1b, patients with 7 selected types of tumor received sugemalimab at the RP2D alone (monotherapy cohorts) or in combination with standard-of-care (SOC) chemotherapy (combination cohorts). The primary endpoint of phase 1b was investigator-assessed objective response rate (ORR). RESULTS: As of 19 February 2020, 29 and 178 patients were treated in phase 1a and 1b, respectively. No dose-limiting toxicities were observed in phase 1a, and the RP2D of sugemalimab was determined as 1200 mg fixed dose once every 3 weeks. Sugemalimab-related adverse events (AEs) were mostly (75.9%) grade 1-2 in phase 1a. Antitumor activity was observed across dose levels with an ORR of 24.1%. In phase 1b, 15.9% and 40.4% of patients in the monotherapy and combination cohorts, respectively, reported grade 3-5 sugemalimab-related AEs. Promising efficacy was observed in all combination cohorts, with ORRs ranging from 47.6 to 75.0%. Exploratory biomarker analysis did not indicate significant differences in responses at different PD-L1 expression/tumor mutation burden levels. CONCLUSIONS: Sugemalimab was well-tolerated and showed promising antitumor activity as monotherapy or in combination with SOC chemotherapy in advanced malignancies. This trial was registered with ClinicalTrials.gov on Oct 18, 2017, number NCT03312842.


Subject(s)
Lymphoma , Neoplasms , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/therapeutic use , Antibodies, Monoclonal, Humanized/adverse effects , Antibodies, Monoclonal, Humanized/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , China , Humans , Immune Checkpoint Inhibitors/adverse effects , Immune Checkpoint Inhibitors/therapeutic use , Lymphoma/drug therapy , Neoplasms/pathology
10.
Eur Radiol ; 32(10): 7295-7306, 2022 10.
Article in English | MEDLINE | ID: mdl-36048205

ABSTRACT

OBJECTIVE: To develop a quantitative Response Evaluation Criteria in Solid Tumors (qRECIST) for evaluating response to neoadjuvant therapy (nT) in ESCCs relying on multiparametric (mp) MRI. METHODS: Patients with cT2-T4a/N0-N3/M0 ESCC undergoing pre-nT and post-nT esophageal mpMRI before radical resection were prospectively included. Images were reviewed by two experienced radiologists. qRECIST was redefined using four methods including conventional criterion (cRECIST) and three model-dependent RECIST relying on quantitative MRI measurements at pre-nT, post-nT, and delta pre-post nT, respectively. Pathological tumor regression grades (TRGs) were used as a reference standard. The rates of agreement between four qRECIST methods and TRGs were determined with a Cronbach's alpha test, area under the curve (AUC), and a diagnostic odds ratio meta-analysis. RESULTS: Ninety-one patients were enrolled. All four methods revealed high inter-reader agreements between the two radiologists, with a Kappa coefficient of 0.96, 0.87, 0.88, and 0.97 for cRECIST, pre-nT RECIST, post-nT RECIST, and delta RECIST, respectively. Among them, delta RECIST achieved the highest overall agreement rate (67.0% [61/91]) with TRGs, followed by post-nT RECIST (63.8% [58/91]), cRECIST (61.5% [56/91]), and pre-nT RECIST (36.3% [33/91]). Especially, delta RECIST achieved the highest accuracy (97.8% [89/91]) in distinguishing responders from non-responders, with 97.3% (34/35) for responders and 98.2% (55/56) for non-responders. Post-nT RECIST achieved the highest accuracy (93.4% [85/91]) in distinguishing complete responders from non-pCRs, with 77.8% (11/18) for pCRs and 94.5% (69/73) for non-pCRs. CONCLUSION: The qRECIST with mpMRI can assess treatment-induced changes and may be used for early prediction of response to nT in ESCC patients. KEY POINTS: • Quantitative mpMRI can reliably assess tumor response, and delta RECIST model had the best performance in evaluating response to nT in ESCCs, with an AUC of 0.98, 0.95, 0.80, and 0.82 for predicting TRG0, TRG1, TRG2, and TRG3, respectively. • In distinguishing responders from non-responders, the rate of agreement between delta RECIST and pathology was 97.3% (34/35) for responders and 98.2% (55/56) for non-responders, resulting in an overall agreement rate of 97.8% (89/91). • In distinguishing pCRs from non-pCR, the rate of agreement between MRI and pathology was 77.8% (11/18) for pCRs and 94.5% (69/73) for non- pCRs, resulting in an overall agreement rate of 91.2% (83/91).


Subject(s)
Esophageal Neoplasms , Esophageal Squamous Cell Carcinoma , Multiparametric Magnetic Resonance Imaging , Esophageal Neoplasms/diagnostic imaging , Esophageal Neoplasms/pathology , Esophageal Neoplasms/therapy , Humans , Neoadjuvant Therapy , Response Evaluation Criteria in Solid Tumors , Treatment Outcome
11.
BMC Gastroenterol ; 21(1): 399, 2021 Oct 23.
Article in English | MEDLINE | ID: mdl-34688262

ABSTRACT

BACKGROUND: Colorectal cancer (CRC) is the third most prevalent cancer worldwide and poses a serious challenge for clinicians. Previous studies have shown promising results in patients with Microsatellite Stable microsatellite-stable CRC refractory to chemotherapy upon treating with (Programmed Cell Death Protein 1) PD-1 inhibitor combined with regorafenib. Herein, we report a unique case of a patient for whom the conventional chemotherapy and radiotherapy were ineffective, but showed a prolonged stable disease with third-line treatment with regorafenib and PD-1 inhibitor, sintilimab. CASE PRESENTATION: A 64-year-old East Asian female patient was admitted to a regional cancer hospital presenting with abdominal unease due to increased stool frequency and bloody stool. Digital anal examination revealed adenocarcinoma, while genetic profiling of the tumor resections detected wild-type KRAS mutations in codon 12 and 13. Microsatellite instability (MSI) analysis for detecting germline mutations of (Mismatch-repair) MMR genes showed stable phenotype. In December 2016, Miles' resection for intestinal adhesion release and iliac vessel exploration in the rectum was performed (Tumor, Node, Metastasis [TNM]: T3N0M0; stage IIA). The adjuvant chemotherapeutic regimen consisted of a combination of capecitabine at 1.5 g (twice daily) and oxaliplatin therapy at 200 mg for three cycles from February 2016; followed by administering capecitabine tablets orally (1.5 g bid) for five cycles as post-operative palliative care. The patient tested positive for hepatic C virus, which was managed by oral antiviral agents. Following recurrence of rectal adenocarcinoma after 4 years and disease progression with a previous chemotherapeutic regimen, regorafenib was administered at 120 mg once daily combined with sintilimab 200 mg, and the patient's progress was monitored. A follow-up computerized tomography imaging in March 2020 showed disease progression, additionally presented nodule formation (TNM: T3NxM1b; stage IVB). According to Response Evaluation Criteria in Solid Tumors criteria (RECIST), the patient showed a complete response (CR) after treatment with regorafenib and sintilimab immunotherapy. CONCLUSION: Data from this clinical case report support future exploration of combination treatment of the oral multi-kinase inhibitor regorafenib with PD-1 targeted monoclonal antibodies in patients with metastatic microsatellite-stable CRC.


Subject(s)
Colorectal Neoplasms , Immune Checkpoint Inhibitors , Antibodies, Monoclonal, Humanized , Antineoplastic Combined Chemotherapy Protocols , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/genetics , Female , Humans , Microsatellite Repeats , Middle Aged , Neoplasm Recurrence, Local , Phenylurea Compounds , Pyridines
12.
Cancer Immunol Immunother ; 69(8): 1493-1504, 2020 Aug.
Article in English | MEDLINE | ID: mdl-32285170

ABSTRACT

Though therapy that promotes anti-tumor response about CD8+ tumor-infiltrating lymphocytes (TILs) has shown great potential, clinical responses to CD8+ TILs immunotherapy vary considerably, largely because of different subpopulation of CD8+ TILs exhibiting different biological characters. To define the relationship between subpopulation of CD8+ TILs and the outcome of antitumor reaction, the phenotype and function of CD103+ CD8+ TILs in esophageal squamous cell carcinoma (ESCC) were investigated. CD103+ CD8+ TILs were presented in ESCC, which displayed phenotype of tissue-resident memory T cells and exhibited high expression of immune checkpoints (PD-1, TIM-3). CD103+ CD8+ TILs were positively associated with the overall survivals of ESCC patients. This population of cells elicited potent proliferation and cytotoxic cytokine secretion potential. In addition, CD103+ CD8+ TILs were elicited potent anti-tumor immunity after anti-PD-1 blockade and were not affected by chemotherapy. This study emphasized the feature of CD103+ CD8+ TILs in immune response and identified potentially new targets in ESCC patients.


Subject(s)
Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Esophageal Neoplasms/immunology , Esophageal Neoplasms/metabolism , Integrin alpha Chains/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Tumor Microenvironment/immunology , 4-Nitroquinoline-1-oxide/toxicity , Adult , Aged , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/immunology , Biomarkers, Tumor , Carcinogens/toxicity , Cohort Studies , Esophageal Neoplasms/chemically induced , Esophageal Neoplasms/pathology , Esophageal Squamous Cell Carcinoma/chemically induced , Esophageal Squamous Cell Carcinoma/immunology , Esophageal Squamous Cell Carcinoma/metabolism , Esophageal Squamous Cell Carcinoma/pathology , Female , Follow-Up Studies , Humans , Integrin alpha Chains/immunology , Male , Mice, Inbred C57BL , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/immunology , Survival Rate , Tumor Cells, Cultured
13.
Biochemistry ; 58(18): 2293-2296, 2019 05 07.
Article in English | MEDLINE | ID: mdl-31021072

ABSTRACT

Blocking the PD-L1/PD-1 interaction with an antibody produces a durable response in patients with diverse advanced cancers. However, it remains elusive on whether the engagement of PD-L1 to PD-1 leads to tumor-intrinsic signaling. In this study, we aim to explore novel protein substrates participating in transducing this tumor-intrinsic PD-L1 signaling. To this end, we performed a BioID (proximity-dependent biotin identification) assay, in which we fused PD-L1 to BirA* (a promiscuous mutant of bacterial biotin ligase BirA) and overexpressed it in the lung adenocarcinoma A549 cell line. Through streptavidin affinity capture and mass spectrometry analysis, we identified 57 candidate proteins including 18 PD-L1/PD-1-interaction-dependent neighbors. In addition to this, 9 out of 57 candidates were involved in the EGFR signaling pathway, which is known to play a critical role in tumorigenesis and multiple therapeutic resistances of lung cancer. This study will provide a new insight in understanding tumor-intrinsic PD-L1-signaling effectors of lung cancer.


Subject(s)
Adenocarcinoma/metabolism , B7-H1 Antigen/metabolism , Biotin/metabolism , Lung Neoplasms/metabolism , Signal Transduction , A549 Cells , Adenocarcinoma/pathology , Carbon-Nitrogen Ligases/genetics , Carbon-Nitrogen Ligases/metabolism , Escherichia coli Proteins/genetics , Escherichia coli Proteins/metabolism , Humans , Lung Neoplasms/pathology , Mass Spectrometry , Mutation , Programmed Cell Death 1 Receptor/metabolism , Protein Binding , Repressor Proteins/genetics , Repressor Proteins/metabolism
14.
Cancer Cell Int ; 19: 322, 2019.
Article in English | MEDLINE | ID: mdl-31827396

ABSTRACT

BACKGROUND: Autologous tumor-infiltrating lymphocytes (Tils) immunotherapy is a promising treatment in patients with advanced hepatocellular cancer. Although Tils treatment has shown great promise, their persistence and the efficacy after adoptive-transfer are insufficient and remain a challenge. Studies have demonstrated that IL-15 and Akt inhibitor can regulate T cell differentiation and memory. Here, we constructed S-15 (Super human IL-15), a fusion protein consisting of human IL-15, the sushi domain of the IL-15 receptor α chain and human IgG-Fc. Herein we compared the effects of S-15 with IL-2 or in combination with Akti on the expansion and activation of Tils. METHODS: Hepatocellular cancer tissues were obtained from 6 patients, Tils were expanded using IL-2, IL-2/S-15, IL-2/Akti or in combination IL-2/S-15/Akti. At day 10, anti-CD3 antibody was added to the culture media and expanded to day 25. The composition, exhaustion and T-cell differentiation markers (CD45RA/CCR7) were analyzed by flow cytometry. RESULTS: We found that IL-2/S-15/Akti expanded Tils and showed the highest percentage of central memory CD45RA-CCR7+ phenotype prior to anti-CD3 antibody activation and after anti-CD3 antibody activation. T cells cultured with IL-2/S-15/Akti exhibited a mixture of CD4+, CD8+, and CD3+CD4-CD8- T cells; S-15 in combination with Akt inhibitor downregulated the expression of PD-1+Tim-3+ on Tils and decreased the Tregs in Tils. Additionally, the Tils expanded in the presence of the Akt inhibitor and S-15 showed enhanced antitumor activity as indicated by the increase in IFN-γ producing tumor infiltrating CD8+ T cells and without comprising the Tils expansion. CONCLUSION: Our study elucidates that IL-2/S-15/Akti expanded Tils and represent a viable source for the cellular therapy for patients with hepatocellular cancer.

15.
Mol Cancer ; 17(1): 73, 2018 03 14.
Article in English | MEDLINE | ID: mdl-29540185

ABSTRACT

BACKGROUND: Liver tumor initiating cells (TICs) have self-renewal and differentiation properties, accounting for tumor initiation, metastasis and drug resistance. Long noncoding RNAs are involved in many physiological and pathological processes, including tumorigenesis. DNA copy number alterations (CNA) participate in tumor formation and progression, while the CNA of lncRNAs and their roles are largely unknown. METHODS: LncRNA CNA was determined by microarray analyses, realtime PCR and DNA FISH. Liver TICs were enriched by surface marker CD133 and oncosphere formation. TIC self-renewal was analyzed by oncosphere formation, tumor initiation and propagation. CRISPRi and ASO were used for lncRNA loss of function. RNA pulldown, western blot and double FISH were used to identify the interaction between lncRNA and CTNNBIP1. RESULTS: Using transcriptome microarray analysis, we identified a frequently amplified long noncoding RNA in liver cancer termed linc00210, which was highly expressed in liver cancer and liver TICs. Linc00210 copy number gain is associated with its high expression in liver cancer and liver TICs. Linc00210 promoted self-renewal and tumor initiating capacity of liver TICs through Wnt/ß-catenin signaling. Linc00210 interacted with CTNNBIP1 and blocked its inhibitory role in Wnt/ß-catenin activation. Linc00210 silencing cells showed enhanced interaction of ß-catenin and CTNNBIP1, and impaired interaction of ß-catenin and TCF/LEF components. We also confirmed linc00210 copy number gain using primary hepatocellular carcinoma (HCC) samples, and found the correlation between linc00210 CNA and Wnt/ß-catenin activation. Of interest, linc00210, CTNNBIP1 and Wnt/ß-catenin signaling targeting can efficiently inhibit tumor growth and progression, and liver TIC propagation. CONCLUSION: With copy-number gain in liver TICs, linc00210 is highly expressed along with liver tumorigenesis. Linc00210 drives the self-renewal and propagation of liver TICs through activating Wnt/ß-catenin signaling. Linc00210 interacts with CTNNBIP1 and blocks the combination between CTNNBIP1 and ß-catenin, driving the activation of Wnt/ß-catenin signaling. Linc00210-CTNNBIP1-Wnt/ß-catenin axis can be targeted for liver TIC elimination.


Subject(s)
Gene Expression Regulation, Neoplastic , Liver Neoplasms/genetics , Liver Neoplasms/metabolism , RNA, Long Noncoding/genetics , Wnt Signaling Pathway , beta Catenin/genetics , beta Catenin/metabolism , Animals , Cell Line, Tumor , Cell Proliferation , DNA Copy Number Variations , Disease Models, Animal , Humans , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Protein Binding , Xenograft Model Antitumor Assays
16.
Biochem Biophys Res Commun ; 496(4): 1169-1175, 2018 02 19.
Article in English | MEDLINE | ID: mdl-29407170

ABSTRACT

The existence of cancer stem cells within the tumor could lead to cancer therapy resistance. TGFß1 is considered as one of the most powerful players in the generation of CSCs through induction of epithelial-mesenchymal transition in different types of cancer including lung cancer, however, the detailed mechanisms by which TGFß1 contribute to EMT induction and CSC maintenance remains unclear. Here, we showed primary lung cancer cells treated by TGFß1 exhibit mesenchymal features, including morphology and expression of mesenchymal marker in a time-dependent manner. We also observed long-term TGFß1 exposure leads to an enrichment of a sub-population of CD44+ CD90+ cells which represent CSCs in lung cancer cells. Moreover, the differential expression microRNAs between CSCs and non-CSCs were identified using next-generation sequencing to screen key miRNAs which might contribute to TGFß1-induced EMT and CSCs generation. Among those differentially expressed miRNAs, the expression of microRNA-138 was time-dependently down-regulated by TGFß1 treatment. We further demonstrated primary lung cancer cells, in which we knockdown the expression of miR-138, exhibit mesenchymal phenotypes and stem cell properties. Taken together, these findings indicate TGFß1-induced down-regulation of microRNA-138 contributes to EMT in primary lung cancer cells, and suggest that miR-138 might serve as a potential therapeutic target.


Subject(s)
Epithelial-Mesenchymal Transition/immunology , Lung Neoplasms/immunology , MicroRNAs/immunology , Neoplastic Stem Cells/immunology , Transforming Growth Factor beta1/immunology , Cell Line, Tumor , Epithelial-Mesenchymal Transition/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/immunology , Humans , Lung Neoplasms/pathology , Neoplastic Stem Cells/drug effects , Transforming Growth Factor beta1/administration & dosage
18.
Cancer Cell Int ; 17: 114, 2017.
Article in English | MEDLINE | ID: mdl-29213216

ABSTRACT

BACKGROUND: T lymphocytes play an indispensably important role in clearing virus and tumor antigen. There is little knowledge about impacts of inhibitory molecules with cytokine on tumor-infiltrating CD4+ T-cells in the presence of gastric cancer (GC). This study investigated the distribution of tumor-infiltrating T-cells subset and the differentiation as well as inhibitory phenotype of T-cells from blood and tissues of GC patients. MATERIALS AND METHODS: Patients with GC diagnosed on the basis of pre-operative staging and laparotomy findings were approached for enrollment between 2014 and 2015 at the Affiliated Cancer Hospital of Zhengzhou University, China. Phenotypic analysis based on isolation of tumor-infiltrating lymphocytes and intracellular IFN-γ staining assay is conducted. Statistical analysis is performed to show significance. RESULTS: The results showed that the percentage of CD4+ T-cells among CD3+ cells in tumors was significantly higher than that in the matched paraneoplastic tissue. CD4+ CD25high CD127low regulatory T-cells (Tregs), PD-1+, Tim-3+, and PD-1+ Tim-3+ cells were up-regulated on tumor infiltrating T-cells from patients with GC compared to their expressions on corresponding peripheral blood and peritumoral T-cells. Blockades of PD-1+ and Tim-3+ were effective in restoring tumor infiltrating T-cells' production of interferon-gamma (IFN-γ). Combined PD-1+ and Tim-3+ inhibition had a synergistic effect on IFN-γ secretion by CD4+ T-cells. CONCLUSION: The results suggested that the composition, inhibitors, and location of the immune infiltrate should be considered when evaluating antitumor immunotherapy. A new insight into the mechanisms underlying T cell dysfunction is provided.

19.
J Surg Oncol ; 114(4): 520-7, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27338835

ABSTRACT

BACKGROUND: NDRG1 plays important roles in tumor growth and metastasis of colorectal cancer (CRC). The relation between NDRG1 and metastatic colorectal cancer (mCRC) has not been identified and the mechanism of NDRG1 involving in mCRC needs to be elucidated. METHODS: Correlations between NDRG1 and clinicopathological characteristics and prognosis of 164 patients with mCRC were evaluated. Sensitivity of NDRG1-knockdown colon cancer cell to irinotecan (CPT-11) was determined by MTT assay. Blocking of NF-κB signaling by p65 siRNA interference was carried out to explore the mechanism of NDRG1 involving in epithelial-mesenchymal transition (EMT)-regulated invasion and metastasis of CRC. RESULTS: NDRG1 expression was significantly negatively correlated with differentiation (P = 0.008) and lymph node metastasis (P = 0.016) of mCRC. NDRG1 was a favorable prognostic factor of mCRC, although might be responsible for CPT-11 resistance in vitro. Knockdown of NDRG1 promoted EMT of CRC cells via NF-κB signaling. Depletion of NDRG1 increased phosphorylation level of NF-κB. E-cadherin expression was increased and Vimentin expression was reduced in the p65-siRNA treated group, compared with the control group (P < 0.001). CONCLUSIONS: NDRG1 appears to prevent EMT-induced metastasis by attenuating NF-κB signaling in mCRC. NDRG1 may be an independent prognostic factor for good survival of mCRC. J. Surg. Oncol. 2016;114:520-527. © 2016 Wiley Periodicals, Inc.


Subject(s)
Cell Cycle Proteins/physiology , Colorectal Neoplasms/pathology , Epithelial-Mesenchymal Transition , Intracellular Signaling Peptides and Proteins/physiology , NF-kappa B/physiology , Signal Transduction/physiology , Adult , Aged , Camptothecin/analogs & derivatives , Camptothecin/pharmacology , Cell Cycle Proteins/genetics , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/mortality , Female , Humans , Intracellular Signaling Peptides and Proteins/genetics , Irinotecan , Lymphatic Metastasis , Male , Middle Aged , Vimentin/analysis
20.
Transl Cancer Res ; 13(5): 2238-2250, 2024 May 31.
Article in English | MEDLINE | ID: mdl-38881916

ABSTRACT

Background: The optional regimens of subsequent therapy after failure of anti-programmed cell death protein-1 (PD-1) antibody in metastatic renal cell carcinoma (mRCC) remain to be explored. There are reports of the efficacy of single-agent vascular endothelial growth factor receptor tyrosine kinase inhibitor (VEGFR-TKI) in patients with mRCC after failure of anti-PD-1 antibody therapy. However, it is not clear whether it is beneficial for patients to receive anti-PD-1 antibody as post-progression treatment. It has great significance to explore whether continuous application of anti-PD-1 antibody is beneficial for patients with mRCC whose diseases progressed to the state of pre-anti-PD-1 therapy. The purposes of this study are to explore the efficacy and safety of subsequent treatment on whether to continue using anti-PD-1 antibody therapy for patients who have progressive mRCC after prior treatment with anti-PD-1 antibody. Methods: The clinical data of patients with mRCC from the Department of Immunotherapy in the Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital from February 1, 2019 to December 31, 2021 were analyzed retrospectively. The primary endpoints were the objective response rate (ORR) and progression-free survival (PFS). The ORR and disease control rate (DCR) were estimated with Fisher's exact test. PFS and overall survival (OS) were assessed using the Kaplan-Meier method and log-rank tests. The associations between potential prognostic variables and PFS were evaluated with univariate and multivariate Cox regression analyses. A P value of less than or equal to 0.05 was deemed as statistically significant. Results: A total of 35 patients were included in this study, during which 19 received VEGFR-TKI monotherapy and 16 received the VEGFR-TKI plus anti-PD-1 antibody therapy. Until the last follow-up on June 30, 2022, 19 patients experienced progressive disease (PD), five were in remission, and 11 kept stable disease (SD). After a median follow-up of 28.7 [95% confidence interval (CI): 17.0-35.6] months, the median PFS (mPFS) was 11.6 months for the VEGFR-TKI group and 9.1 months for the VEGFR-TKI plus anti-PD-1 antibody group [hazard ratio (HR) =0.81, 95% CI: 0.32-1.03, P=0.44]. Median OS (mOS) were 16.9 and 11.2 months respectively (HR =0.99, 95% CI: 0.44-2.27, P=0.90). The ORRs were 26.3% and 0% (P=0.049), and the DCRs were 47.4% and 43.8% (P=0.55) respectively. Treatment-related adverse events (TRAEs) occurred in 14 patients (73.7%) in the VEGFR-TKI group and 14 patients (87.5%) in the VEGFR-TKI plus anti-PD-1 antibody group (P=0.42); grade 3/4 TRAEs occurred in two patients (10.5%) and six patients (37.5%) respectively (P=0.11). Conclusions: VEGFR-TKI monotherapy is an efficacious regimen for patients with mRCC whose diseases progressed on previous anti-PD-1 antibody therapy, and continuous anti-PD-1 therapy after failure of anti-PD-1 antibody could not provide additional clinical benefit but increased the incidence of TRAEs.

SELECTION OF CITATIONS
SEARCH DETAIL