Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 227
Filter
1.
Support Care Cancer ; 32(6): 370, 2024 May 22.
Article in English | MEDLINE | ID: mdl-38776028

ABSTRACT

OBJECTIVE: Immune checkpoint inhibitors (ICIs), specifically targeting the programmed cell death protein-1 or its ligand (PD-1/PD-L1), have been extensively used in the treatment of a spectrum of malignancies, although the predictive biomarkers remain to be elucidated. This study aims to investigate the association between baseline circulating levels of cytokines and the creatinine/cystatin C ratio (CCR) with the treatment outcomes of ICIs in patients with advanced cancer. METHODS: The pre-treatment circulating levels of 10 cytokines (PD-L1, CTLA4, CXCL10, LAG3, HGF, CCL2, MIG, GRANB, IL-18, and IL-6) were measured via automated capillary-based immunoassay platform in the serum of 65 advanced cancer patients treated with anti-PD-1/PD-L1-based systemic therapy and 10 healthy volunteers. The levels of cytokines and CCR were quantified and categorized into high and low groups based on the median value. The associations of serum cytokines and CCR with response to treatment, survival, and immune-related adverse events were assessed. RESULTS: Elevated circulating levels of 6 cytokines (PD-L1, CXCL10, HGF, CCL2, MIG, and IL-6) were observed in cancer patients compared with that in healthy volunteers. The correlation coefficients between cytokines, CCR and nutritional risk index were also calculated. In the cancer cohort (N = 65), low circulating HGF (P = 0.023, P = 0.029), low IL-6 (P = 0.002, P < 0.001), and high CCR (P = 0.031, P = 0.008) were associated with significantly improved progression-free survival (PFS) and overall survival (OS). Multi-variable COX analyses adjusted for clinicopathological factors revealed that low HGF, low IL-6, and high CCR were independent favorable prognostic factors for PFS (P = 0.028, P = 0.010, and P = 0.015, respectively) and OS (P = 0.043, P = 0.003, and P = 0.026, respectively). Grade 2 irAEs occurred more frequently in patients with low levels of circulating CCL2 and LAG3. CONCLUSIONS: Pre-treatment circulating levels of serum IL-6, HGF, and CCR may serve as independent predictive and prognostic biomarkers in advanced cancer patients treated with ICIs-based systemic therapy. These findings might help to identify potential patients who would benefit from these therapies.


Subject(s)
Biomarkers, Tumor , Creatinine , Cytokines , Immune Checkpoint Inhibitors , Neoplasms , Humans , Male , Female , Neoplasms/drug therapy , Neoplasms/blood , Middle Aged , Aged , Cytokines/blood , Prognosis , Immune Checkpoint Inhibitors/therapeutic use , Immune Checkpoint Inhibitors/pharmacology , Creatinine/blood , Biomarkers, Tumor/blood , Adult , Aged, 80 and over , B7-H1 Antigen/blood , Case-Control Studies
2.
Front Immunol ; 15: 1370771, 2024.
Article in English | MEDLINE | ID: mdl-38707906

ABSTRACT

Introduction: Anti-PD-1/PD-L1 inhibitors therapy has become a promising treatment for hepatocellular carcinoma (HCC), while the therapeutic efficacy varies significantly among effects for individual patients are significant difference. Unfortunately, specific predictive biomarkers indicating the degree of benefit for patients and thus guiding the selection of suitable candidates for immune therapy remain elusive.no specific predictive biomarkers are available indicating the degree of benefit for patients and thus screening the preferred population suitable for the immune therapy. Methods: Ultra-high-pressure liquid chromatography-mass spectrometry (UHPLC-MS) considered is an important method for analyzing biological samples, since it has the advantages of high rapid, high sensitivity, and high specificity. Ultra-high-pressure liquid chromatography-mass spectrometry (UHPLC-MS) has emerged as a pivotal method for analyzing biological samples due to its inherent advantages of rapidity, sensitivity, and specificity. In this study, potential metabolite biomarkers that can predict the therapeutic effect of HCC patients receiving immune therapy were identified by UHPLC-MS. Results: A partial least-squares discriminant analysis (PLS-DA) model was established using 14 glycerophospholipid metabolites mentioned above, and good prediction parameters (R2 = 0.823, Q2 = 0.615, prediction accuracy = 0.880 and p < 0.001) were obtained. The relative abundance of glycerophospholipid metabolite ions is closely related to the survival benefit of HCC patients who received immune therapy. Discussion: This study reveals that glycerophospholipid metabolites play a crucial role in predicting the efficacy of immune therapy for HCC.


Subject(s)
B7-H1 Antigen , Biomarkers, Tumor , Carcinoma, Hepatocellular , Immune Checkpoint Inhibitors , Liver Neoplasms , Carcinoma, Hepatocellular/drug therapy , Carcinoma, Hepatocellular/blood , Carcinoma, Hepatocellular/immunology , Humans , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/blood , Chromatography, High Pressure Liquid/methods , Male , Immune Checkpoint Inhibitors/therapeutic use , Biomarkers, Tumor/blood , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/blood , Female , Middle Aged , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Mass Spectrometry/methods , Aged , Metabolomics/methods , Glycerophospholipids/blood
3.
Crit Rev Oncol Hematol ; 197: 104332, 2024 May.
Article in English | MEDLINE | ID: mdl-38580184

ABSTRACT

Immune checkpoints inhibitors (ICIs) have markedly improved the therapeutic management of advanced NSCLC and, more recently, they have demonstrated efficacy also in the early-stage disease. Despite better survival outcomes with ICIs compared to standard chemotherapy, a large proportion of patients can derive limited clinical benefit from these agents. So far, few predictive biomarkers, including the programmed death-ligand 1 (PD-L1), have been introduced in clinical practice. Therefore, there is an urgent need to identify novel biomarkers to select patients for immunotherapy, to improve efficacy and avoid unnecessary toxicity. A deeper understanding of the mechanisms involved in antitumor immunity and advances in the field of liquid biopsy have led to the identification of a wide range of circulating biomarkers that could potentially predict response to immunotherapy. Herein, we provide an updated overview of these circulating biomarkers, focusing on emerging data from clinical studies and describing modern technologies used for their detection.


Subject(s)
Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung , Immune Checkpoint Inhibitors , Lung Neoplasms , Humans , Immune Checkpoint Inhibitors/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/diagnosis , Biomarkers, Tumor/blood , Lung Neoplasms/drug therapy , Lung Neoplasms/blood , Lung Neoplasms/diagnosis , Lung Neoplasms/pathology , Immunotherapy/methods , Prognosis , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/blood
4.
Anticancer Drugs ; 35(5): 418-425, 2024 Jun 01.
Article in English | MEDLINE | ID: mdl-38386011

ABSTRACT

The clinical significance of plasma soluble programmed cell death ligand 1 (sPD-L1) and vascular endothelial growth factor (VEGF) for non-small cell lung cancer (NSCLC) treated with the combination of anti-angiogenic therapy and anti-PD-L1 antibody (Ab) remain unknown. This study aimed to explore the association between plasma sPD-L1 and VEGF levels and the prognosis of NSCLC patients treated with the combination of Envafolimab and Endostar. Peripheral blood samples were collected from 24 NSCLC patients at baseline and after 6 weeks of treatment and were detected for sPD-L1 and VEGF levels. Both baseline and posttreatment sPD-L1 were significantly higher in progressive disease (PD) group than in controlled disease (CD) group (median: 77.5 pg/ml vs. 64.6 pg/ml, P  = 0.036, median: 8451 pg/ml vs. 5563 pg/ml, P  = 0.012). In multivariate analysis, lower baseline sPD-L1 levels were significantly associated with longer progression-free survival (PFS) (HR = 6.834, 95% CI: 1.350-34.592, P  = 0.020). There were significantly higher posttreatment VEGF levels in PD group compared with CD group (median: 323.7 pg/ml vs. 178.5 pg/ml, P  = 0.009). Higher posttreatment VEGF levels were significantly associated with shorter PFS in multivariate analysis (HR = 5.911, 95% CI: 1.391-25.122, P  = 0.016). Plasma sPD-L1 and VEGF levels are associated with the clinical response and prognosis of NSCLC patients treated with the combination of PD-L1 inhibitors and anti-angiogenetic therapy.


Subject(s)
B7-H1 Antigen , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Vascular Endothelial Growth Factor A , Humans , Biomarkers, Tumor , Carcinoma, Non-Small-Cell Lung/diagnosis , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy , Lung Neoplasms/drug therapy , Prognosis , Vascular Endothelial Growth Factor A/blood , Vascular Endothelial Growth Factor A/chemistry , B7-H1 Antigen/blood , B7-H1 Antigen/chemistry
5.
Tumour Biol ; 46(s1): S355-S367, 2024.
Article in English | MEDLINE | ID: mdl-38277316

ABSTRACT

BACKGROUND: Programmed cell death receptors and ligands in cancer tissue samples are established companion diagnostics for immune checkpoint inhibitor (ICI) therapies. OBJECTIVE: To investigate the relevance of soluble PD-1, PD-L1 and PD-L2 for estimating therapy response and prognosis in non-small cell lung cancer patients (NSCLC) undergoing platin-based combination chemotherapies. METHODS: In a biomarker substudy of a prospective, multicentric clinical trial (CEPAC-TDM) on advanced NSCLC patients, soluble PD-1, PD-L1 and PD-L2 were assessed in serial serum samples by highly sensitive enzyme-linked immunosorbent assays and correlated with radiological response after two cycles of chemotherapy and with overall survival (OS). RESULTS: Among 243 NSCLC patients, 185 achieved response (partial remission and stable disease) and 58 non-response (progression). The distribution of PD-1, PD-L1 and PD-L2 at baseline (C1), prior to staging (C3) and the relative changes (C3/C1) greatly overlapped between the patient groups with response and non-response, thus hindering the discrimination between the two groups. None of the PD markers had prognostic value regarding OS. CONCLUSIONS: Neither soluble PD-1, PD-L1 nor PD-L2 did provide clinical utility for predicting response to chemotherapy and prognosis. Studies on the relevance of PD markers in ICI therapies are warranted.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , B7-H1 Antigen/blood , B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/metabolism , Lung Neoplasms/drug therapy , Lung Neoplasms/metabolism , Prognosis , Programmed Cell Death 1 Receptor/blood , Prospective Studies
6.
Front Immunol ; 14: 1325462, 2023.
Article in English | MEDLINE | ID: mdl-38149256

ABSTRACT

Introduction: The clinical relevance of soluble forms of programmed cell death-1 (sPD-1) and programmed cell death-ligand 1 (sPD-L1) remains unclear. We here investigated the relation between the efficacy of PD-1 blockade and pretreatment plasma levels of sPD-1 and sPD-L1 across a broad range of cancer types. Methods: We retrospectively analyzed clinical data from 171 patients with advanced solid tumors who received nivolumab or pembrolizumab monotherapy regardless of treatment line. The concentrations of sPD-1 and sPD-L1 were measured with a fully automated immunoassay (HISCL system). Results: The study subjects comprised patients with head and neck cancer (n = 50), urothelial cancer (n = 42), renal cell cancer (n = 37), gastric cancer (n = 20), esophageal cancer (n = 10), malignant pleural mesothelioma (n = 6), or microsatellite instability-high tumors (n = 6). High or low levels of sPD-1 or sPD-L1 were not significantly associated with progression-free survival (PFS) or overall survival (OS) for PD-1 blockade in the entire study population. Comparison of treatment outcomes according to combinations of high or low sPD-1 and sPD-L1 levels, however, revealed that patients with low sPD-1 and high sPD-L1 concentrations had a significantly poorer PFS (HR of 1.79 [95% CI, 1.13-2.83], p = 0.01) and a tendency toward poorer OS (HR of 1.70 [95% CI, 0.99-2.91], p = 0.05) compared with all other patients. Conclusion: Our findings suggest that the combination of low sPD-1 and high sPD-L1 levels is a potential negative biomarker for PD-1 blockade therapy.


Subject(s)
Neoplasms , Programmed Cell Death 1 Receptor , Retrospective Studies , Humans , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Programmed Cell Death 1 Receptor/blood , Neoplasms/blood , Neoplasms/drug therapy , Neoplasms/pathology , B7-H1 Antigen/blood , Male , Female , Treatment Outcome , Antibodies, Monoclonal/therapeutic use , Nivolumab/therapeutic use , Immune Checkpoint Inhibitors/therapeutic use
7.
Immunol Res ; 70(5): 698-707, 2022 10.
Article in English | MEDLINE | ID: mdl-35732880

ABSTRACT

Sepsis causes a myriad of immunological reactions that result in life-threatening alterations in the human body. Immunosuppression in sepsis is partly attributed to the programmed death receptor (PD-1) and its associated ligand (PD-L1) via the regulation of lymphocytes and neutrophils. Although the soluble forms of these proteins (i.e., sPD-1 and sPD-L1, respectively) are recognized as possible sepsis biomarkers, their functional implications are yet to be elucidated. Our research assessed the correlation between sPD-1 and sPD-L1 and blood mRNA markers and sepsis outcome. Blood samples of septic patients of urogenital origin versus control patients (both groups: n = 18) were analyzed. Blood serum sPD-1 and sPD-L1 levels were determined using the enzyme-linked immunosorbent assay (ELISA). The whole blood mRNA concentrations of PD-1, PD-L1, neutrophil markers (CEACAM8 and MPO), and T-lymphocyte markers (TCRß, CD4 and CD8) were determined via reverse transcriptase quantitative PCR (RT-qPCR). sPD-L1 levels were significantly increased in septic patients when compared to the controls, whereas sPD-1 levels were unaltered. Patients with high sPD-L1 levels, as dichotomized to the median, had a significantly shorter survival rate than those with low sPD-L1 levels. The sensitivity/specificity characteristics of sPD-L1 proved significant for sepsis detection. Furthermore, sPD-L1 correlated with the mRNA concentrations of PD-L1, CEACAM, and MPO, as well as major inflammatory markers (C-reactive protein and procalcitonin). However, sPD-L1 negatively correlated with TCRß, CD4, and CD8 mRNAs. sPD-L1 was found to be significantly increased in septic patients. Notably, sPD-L1 correlated with PD-L1 mRNA and neutrophil markers and was indicative of adverse outcomes.


Subject(s)
B7-H1 Antigen , Lymphocytes , Neutrophils , Sepsis , B7-H1 Antigen/blood , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Biomarkers/blood , C-Reactive Protein , Humans , Ligands , Lymphocytes/immunology , Neutrophils/immunology , Procalcitonin , Prognosis , Programmed Cell Death 1 Receptor/genetics , RNA, Messenger/genetics , RNA, Messenger/immunology , RNA-Directed DNA Polymerase , Receptors, Death Domain , Sepsis/blood , Sepsis/genetics , Sepsis/immunology
8.
Int J Oncol ; 60(4)2022 04.
Article in English | MEDLINE | ID: mdl-35211760

ABSTRACT

Anti­programmed death­1 (PD­1)/programmed death­ligand 1 (PD­L1)­directed immunotherapy has revolutionized the treatment of advanced non­small cell lung cancer (NSCLC). However, predictive biomarkers are still lacking, particularly in identifying PD­L1low/negative patients who will benefit from immunotherapy. It was previously reported that farnesoid X receptor (FXR) downregulated PD­L1 expression in NSCLC, and that FXRhighPD­L1low mouse Lewis lung carcinoma tumors showed an increased susceptibility to PD­1 blockade compared with mock tumors. At present, whether the FXRhighPD­L1low phenotype predicts clinical response to immunotherapy in patients with NSCLC remains unclear. Herein, a retrospective study was conducted to examine the expression levels of FXR, PD­L1 and CD8+ T cells by immunohistochemistry in a cohort of 149 patients with NSCLC receiving anti­PD­1­based chemo­immunotherapy. The results revealed that high FXR and PD­L1 expression levels were associated with higher objective response rates (ORR) in all patients. High PD­L1 expression also indicated superior progression­free survival (PFS). Interestingly, an inverse correlation was identified between FXR and PD­L1 expression in specimens with NSCLC. Subgroup analysis revealed that high FXR expression was associated with a higher ORR, as well as longer PFS and overall survival (OS) in PD­L1low patients. Cox multivariate analysis revealed that high FXR expression was an independent predictor for PFS and OS in PD­L1low patients. Tumor microenvironment evaluation revealed a statistically significant decrease of infiltrating CD8+ T cells in FXRhigh specimens with NSCLC. Overall, the present study proposed an FXRhighPD­L1low signature as a candidate predictor of response to anti­PD­1­based chemo­immunotherapy in PD­L1low/negative patients with NSCLC, providing evidence that could be used to broaden the patients benefitting from immunotherapy.


Subject(s)
B7-H1 Antigen/analysis , Carcinoma, Non-Small-Cell Lung/complications , Predictive Value of Tests , Receptors, Cytoplasmic and Nuclear/analysis , Adult , Aged , B7-H1 Antigen/blood , Biomarkers, Tumor/analysis , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/physiopathology , Drug Therapy/methods , Drug Therapy/statistics & numerical data , Female , Humans , Immunotherapy/methods , Immunotherapy/statistics & numerical data , Male , Middle Aged , Receptors, Cytoplasmic and Nuclear/blood , Receptors, Cytoplasmic and Nuclear/metabolism , Survival Analysis
9.
Klin Lab Diagn ; 67(2): 76-80, 2022 Feb 23.
Article in English | MEDLINE | ID: mdl-35192751

ABSTRACT

Results of enzyme-linked immunosorbent assay of the soluble forms of PD-1/PD-L immune checkpoint receptor and ligand (sPD-1 and sPD-L1) in pretreatment blood serum of 88 breast cancer patients at various disease stages aged 30-83 years are presented. The control group included 55 practically healthy women aged 19-82 years. Serum sPD-1 and sPD-L1 levels in breast cancer patients highly significantly (p<0.0001) differ from control and these changes are opposite: soluble receptor level is more than 6-fold decreased, while soluble ligand concentration - 5.5 fold increased. Both markers separately, as well as their ratio demonstrate very high sensitivity (94-100%) and specificity (95-100%) in relation to healthy control. No statistically significant associations of sPD-1 and sPD-L1 levels with clinical stage, individual TNM system criteria, tumor histological structure, grade, receptor status, and molecular type were established. In particular, no significant peculiarities of the markers' levels in triple negative breast cancer successfully treated with anti-PD-1/PD-L1 preparations were revealed. Long-term follow-up and dynamic studies of sPD-1 and sPD-L1serum levels in the course of treatment are required for evaluation of their independent from clinical and morphological factors prognostic significance and the possibility of application as low invasive tests for prediction and monitoring of corresponding targeted therapy efficiency.


Subject(s)
B7-H1 Antigen , Breast Neoplasms , Programmed Cell Death 1 Receptor , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/blood , Breast Neoplasms/blood , Breast Neoplasms/genetics , Female , Humans , Ligands , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/blood , Serum , Young Adult
10.
J Immunol ; 208(5): 1248-1258, 2022 03 01.
Article in English | MEDLINE | ID: mdl-35173033

ABSTRACT

Paroxysmal nocturnal hemoglobinuria (PNH) is a rare hemolytic disease driven by impaired complement regulation. Mutations in genes encoding the enzymes that build the GPI anchors are causative, with somatic mutations in the PIG-A gene occurring most frequently. As a result, the important membrane-bound complement regulators CD55 and CD59 are missing on the affected hematopoietic stem cells and their progeny, rendering those cells vulnerable to complement attack. Immune escape mechanisms sparing affected PNH stem cells from removal are suspected in the PNH pathogenesis, but molecular mechanisms have not been elucidated. We hypothesized that exuberant complement activity in PNH results in enhanced immune checkpoint interactions, providing a molecular basis for the potential immune escape in PNH. In a series of PNH patients, we found increased expression levels of the checkpoint ligand programmed death-ligand 1 (PD-L1) on granulocytes and monocytes, as well as in the plasma of PNH patients. Mechanistically, we demonstrate that complement activation leading to the decoration of particles/cells with C3- and/or C4-opsonins increased PD-L1 expression on neutrophils and monocytes as shown for different in vitro models of classical or alternative pathway activation. We further establish in vitro that complement inhibition at the level of C3, but not C5, inhibits the alternative pathway-mediated upregulation of PD-L1 and show by means of soluble PD-L1 that this observation translates into the clinical situation when PNH patients are treated with either C3 or C5 inhibitors. Together, the presented data show that the checkpoint ligand PD-L1 is increased in PNH patients, which correlates with proximal complement activation.


Subject(s)
B7-H1 Antigen/metabolism , Complement Activation/immunology , Complement C3/antagonists & inhibitors , Complement C5/antagonists & inhibitors , Hemoglobinuria, Paroxysmal/pathology , B7-H1 Antigen/blood , CD55 Antigens/genetics , CD59 Antigens/genetics , Complement C3/immunology , Complement C5/immunology , Granulocytes/metabolism , Hematopoietic Stem Cells/cytology , Hemoglobinuria, Paroxysmal/immunology , Humans , Immune Evasion/immunology , Membrane Proteins/genetics , Monocytes/metabolism
11.
Comput Math Methods Med ; 2022: 8375349, 2022.
Article in English | MEDLINE | ID: mdl-35126639

ABSTRACT

Background soluble programmed death-ligand 1 (sPD-L1) expression in lung squamous cell carcinoma and lung adenocarcinoma is associated with disease progression, and sPD-L1 expression in small cell lung cancer (SCLC) may have similar manifestations and become a potential marker for treatment. The purpose of this study was to observe the changes of plasma sPD-L1 expression in SCLC patients. Methods. 90 patients diagnosed with SCLC from January 2019 to November 2020 were selected as the test group, including 72 males and 18 women, 58.7 ± 6.6 years; 30 healthy subjects were selected from the physical examination center, including 18 males, 12 females, and 60.3 ± 7.0 years. There were no statistical difference in sex and age factors between the trial and control groups (p > 0.05). Selected SCLC used chemotherapy regimen: cisplatin + etoposide (EP), carboplatin + etoposide (CE), and SCLC group were divided into three subgroups of disease progression group, partial remission group, and disease stability group according to the treatment effect. Comparison of the differences in sPD-L1 expression content between the experimental and control populations. Plasma sPD-L1 levels were dynamically monitored pre- and posttreatment in 90 patients with small-cell lung cancer and were associated with efficacy among subgroups. Meanwhile, the risk factors for patient sPD-L1 expression content were analyzed by logistic regression. Results. Plasma sPD-L1 levels were higher in the SCLC group than in the healthy people group (t = 7.40, p < 0.01). In the disease progression group of the SCLC group, sPD-L1 levels were decreased in the SCLC group, sPD-L1 in some remission group was increased after treatment, and sPD-L1 levels in the disease-stable group (p > 0.05). Multivariate logistic regression analysis showed that factors promoting increased sPD-L1 expression in SCLC patients included increased smoking, brain metastasis, and ProGRP expression (both p values < 0.05). Conclusion. (1) Higher peripheral sPD-L1 expression in SCLC patients than in healthy patients, and the expression levels were closely related to efficacy. (2) Dynamic changes in s PD-L1 were correlated with clinical efficacy. (3) The progression of sPD-L1 and ProGRP in SCLC patients showed the same extent during remission and stabilization, suggesting the effect of s PD-L1 in the evaluation of SCLC tumors and the reflection of the tumor marker ProGRP.


Subject(s)
B7-H1 Antigen/blood , Lung Neoplasms/blood supply , Small Cell Lung Carcinoma/blood , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/blood , Case-Control Studies , Computational Biology , Female , Humans , Logistic Models , Lung Neoplasms/drug therapy , Male , Middle Aged , Risk Factors , Small Cell Lung Carcinoma/drug therapy , Solubility , Treatment Outcome
12.
Anticancer Res ; 42(2): 801-810, 2022 Feb.
Article in English | MEDLINE | ID: mdl-35093878

ABSTRACT

BACKGROUND/AIM: Current treatment strategies for advanced melanoma require serial assessment of disease status in affected patients. In this study, we sought to examine the relationship between radiographic tumour burden and blood borne biomarkers including plasma cfDNA, serum LDH, plasma VEGF, PD-L1 and IFN-γ in advanced melanoma patients receiving immunotherapy. We hypothesized that a combination of these explanatory variables in a suitable regression analysis model may predict changes in tumour burden during patient treatment. MATERIALS AND METHODS: We extracted and quantified circulating cfDNA, LDH, VEGF, PD-L1, and IFN-γ from thirty patients with stage IV melanoma at baseline and at six months. All participating patients were evaluated with paired blood sample collection and CT scan assessments during treatment. RESULTS: Changes in radiographic tumour burden correlated with changes in levels of cfDNA (p≤0.001), LDH (p≤0.001), VEGF (p≤0.001), and PD-L1 (p<0.05) during treatment. Multiple regression analysis consisting of the follow-up to baseline assessment ratios of cfDNA, LDH, VEGF and PD-L1 explained changes in tumour burden (F (4, 23)=32.05, p<0.001); with an R2 of 0.8479 (Y=ß0+ß1*B+ß2*C+ß3*D+ß4*E). CONCLUSION: A quantitative measure of cfDNA, LDH, VEGF and PD-L1 may complement current methods of assessing tumour burden in advanced melanoma patients.


Subject(s)
Melanoma/blood , Melanoma/therapy , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/blood , Biomarkers, Tumor/blood , Cell-Free Nucleic Acids/blood , Female , Humans , Immunotherapy , Interferon-gamma/blood , L-Lactate Dehydrogenase/blood , Male , Melanoma/pathology , Middle Aged , Regression Analysis , Tumor Burden , Vascular Endothelial Growth Factor A/blood
13.
J Pathol Clin Res ; 8(3): 257-267, 2022 05.
Article in English | MEDLINE | ID: mdl-35037417

ABSTRACT

Immunotherapy targeting programmed cell death-1 (PD-1) has considerably improved the prognosis of patients with advanced cancers; however, its efficacy in the treatment of pancreatic ductal adenocarcinoma (PDAC) is unfavourable. To address the issue of PDAC immunotherapy, we investigated the expression of two PD-1 ligands, PD-L1 and PD-L2, in PDAC, analysed their role in survival, and explored their correlation with clinicopathological features, immune infiltration, and DNA damage response molecules. Immunohistochemistry was performed on 291 surgically resected PDAC samples. In tumour cells (TCs) and immune cells (ICs), the positivity of PD-L1 expression was 30 and 20% and that of PD-L2 expression was 40 and 20%, respectively. Moreover, PD-L1 expression on TCs correlated with its expression on ICs (p < 0.0001); a similar result was observed for PD-L2 (p < 0.0001). Nonetheless, no correlation was observed between PD-L1 and PD-L2 expression. Positive PD-L1 expression on TCs was related to N1 stage (p = 0.011) and AJCC II stage (p = 0.002), whereas positive PD-L2 expression on TCs was associated with high FOXP3+ cell infiltration (p = 0.001) and high BRCA2 expression (p < 0.0001). Survival analysis revealed that positive PD-L1 (p = 0.046) and PD-L2 (p = 0.028) expression on TCs was an independent risk factor for unfavourable disease-specific survival (DSS). Furthermore, positive PD-L2 expression on TCs was an independent risk factor for lower DSS in the pN0 (p = 0.023), moderate and well tumour differentiation (p = 0.004), low BRCA1 (p = 0.017), wild-type p53 (p = 0.034), and proficient mismatch repair (p = 0.004) subgroups. Moreover, post-operative adjuvant chemotherapy could significantly affect DSS, regardless of PD-L1/PD-L2 expression status (positive or negative) on TCs, while it only prolonged DSS in PDL1-ICs(-) (p < 0.0001) and PDL2-ICs(-) (p < 0.0001) subgroups. This study provides a comprehensive understanding of the roles of PD-L1 and PD-L2 in PDAC, supporting anti-PD-1 axis immunotherapy for PDAC.


Subject(s)
B7-H1 Antigen , Carcinoma, Pancreatic Ductal , DNA Damage , Lymphocytes, Tumor-Infiltrating , Pancreatic Neoplasms , Programmed Cell Death 1 Ligand 2 Protein , B7-H1 Antigen/biosynthesis , B7-H1 Antigen/blood , B7-H1 Antigen/genetics , B7-H1 Antigen/immunology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/immunology , Carcinoma, Pancreatic Ductal/pathology , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/immunology , Pancreatic Neoplasms/pathology , Programmed Cell Death 1 Ligand 2 Protein/biosynthesis , Programmed Cell Death 1 Ligand 2 Protein/genetics , Programmed Cell Death 1 Ligand 2 Protein/immunology
14.
Oncology ; 100(1): 39-47, 2022.
Article in English | MEDLINE | ID: mdl-34991094

ABSTRACT

INTRODUCTION: We determined the soluble programmed cell death-1 ligand-1 (sPD-L1) concentration in patients with esophageal squamous cell carcinoma (ESCC), and confirmed the PD-L1 expression in resected specimens. METHODS: Blood samples were collected from 73 patients with histologically proven ESCC. The serum levels of sPD-L1 were measured using an enzyme-linked immunosorbent assay. The correlations between the sPD-L1 concentration and the expression of PD-L1 in tumor specimens and tumor depth, lymph node metastasis, disease stage, and various laboratory data were assessed. RESULTS: sPD-L1 levels in patients with high PD-L1 expression levels in tumor tissue were significantly higher than in patients with low PD-L1 expression levels (p = 0.042). The OS of the sPD-L1-high group was significantly worse than that of the low group (p = 0.028). Similarly, patients in whom a tissue specimen was PD-L1-positive group showed significantly poorer OS. CONCLUSION: The sPD-L1 concentration was correlated with the PD-L1 expression in tissues. Patients with PD-L1-positive tissue specimens showed significantly higher sPD-L1 levels in comparison to PD-L1-negative cases. Furthermore, patients with high sPD-L1 expression levels had a significantly worse prognosis than those with low sPD-L1 expression levels, and patients with a PD-L1-positive tissue specimen had a significantly worse prognosis than patients in whom the tissue specimen showed a low PD-L1 expression level.


Subject(s)
B7-H1 Antigen/metabolism , Biomarkers, Tumor/metabolism , Esophageal Neoplasms/metabolism , Esophageal Squamous Cell Carcinoma/metabolism , Aged , Animals , B7-H1 Antigen/blood , B7-H1 Antigen/genetics , Biomarkers, Tumor/blood , Cell Line, Tumor , Esophageal Neoplasms/mortality , Esophageal Neoplasms/pathology , Esophageal Neoplasms/surgery , Esophageal Squamous Cell Carcinoma/mortality , Esophageal Squamous Cell Carcinoma/pathology , Esophageal Squamous Cell Carcinoma/surgery , Female , Humans , Male , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Prognosis , Solubility , Survival Rate , Xenograft Model Antitumor Assays
15.
Gene ; 806: 145935, 2022 Jan 05.
Article in English | MEDLINE | ID: mdl-34478821

ABSTRACT

Soluble molecules of programmed death ligand 1 (sPD-L1) are known to modulate T-cell depletion, an important mechanism of hepatitis B virus (HBV) persistence and liver disease progression. In addition, PD-L1 polymorphisms in the 3'-UTR can influence PD-L1 expression and have been associated with cancer risk, although not definitively. The purpose of this study was to investigate the association of PD-L1 polymorphisms and circulating levels of sPD-L1 in HBV infection and live disease progression. In this study, five hundred fifty-one HBV infected patients of the three clinically well-defined subgroups chronic hepatitis B (CHB, n = 186), liver cirrhosis (LC, n = 142) and hepatocellular carcinoma (HCC, n = 223) and 240 healthy individuals (HC) were enrolled. PD-L1 polymorphisms (rs2297136 and rs4143815) were genotyped by in-house validated ARMS assays. Logistic regression models were applied in order to determine the association of PD-L1 polymorphisms with HBV infection as well as with progression of related liver diseases. Plasma sPD-L1 levels were quantified by ELISA assays. The PD-L1 rs2297136 AA genotype was associated with HBV infection susceptibility (HBV vs. HC: OR = 1.6; 95%CI = 1.1-2.3; p = 0.0087) and disease progression (LC vs. CHB: OR = 1.8; 95%CI = 1.1-2.9; p = 0.018). Whereas, the rs2297136 GG genotype was a protective factor for HCC development. Plasma sPD-L1 levels were significantly high in HBV patients (p < 0.0001) and higher in the LC followed by CHB and HCC groups. High sPD-L1 levels correlated with increased liver enzymes and with advanced liver disease progression (Child-pugh C > B > A, p < 0.0001) and BCLC classification (BCLC D > C > B > A, p = 0.031). We could, for the first time, conclude that PD-L1 rs2297136 polymorphism and plasma sPD-L1 protein levels associate with HBV infection and HBV-related liver disease progression.


Subject(s)
B7-H1 Antigen/genetics , Carcinoma, Hepatocellular/genetics , Hepatitis B virus/pathogenicity , Hepatitis B, Chronic/genetics , Liver Cirrhosis/genetics , Liver Neoplasms/genetics , Polymorphism, Genetic , 3' Untranslated Regions , Adult , Aged , B7-H1 Antigen/blood , Biomarkers/blood , Carcinoma, Hepatocellular/diagnosis , Carcinoma, Hepatocellular/etiology , Carcinoma, Hepatocellular/virology , Case-Control Studies , Disease Progression , Female , Gene Expression , Genetic Predisposition to Disease , Hepatitis B virus/growth & development , Hepatitis B, Chronic/complications , Hepatitis B, Chronic/diagnosis , Hepatitis B, Chronic/virology , Humans , Liver/metabolism , Liver/pathology , Liver/virology , Liver Cirrhosis/diagnosis , Liver Cirrhosis/etiology , Liver Cirrhosis/virology , Liver Neoplasms/diagnosis , Liver Neoplasms/etiology , Liver Neoplasms/virology , Male , Middle Aged , Predictive Value of Tests
16.
Cancer Immunol Immunother ; 71(3): 661-674, 2022 Mar.
Article in English | MEDLINE | ID: mdl-34322779

ABSTRACT

BACKGROUND: Exosomal PD-L1 (exoPD-L1) could induce immunosuppression functionally, thus impairing patients' survival in melanoma, NSCLC, and gastric cancer. However, no evidence demonstrates the feasibility of circulating exoPD-L1 and soluble PD-L1 (sPD-L1) as biomarkers for prognosis and early recurrence in colorectal liver metastasis (CRLM) patients following hepatectomy or their association with T cell infiltration at liver metastases. METHODS: In cohort 1, exoPD-L1 and sPD-L1 were preoperatively tested using ELISA. CD3, CD8, granzyme B (GB) and PD1 expressed at liver metastases were evaluated using immunohistochemistry. In cohort 2, exoPD-L1 and sPD-L1 were detected at baseline, before hepatectomy, after hepatectomy, and after disease progression. RESULTS: In cohort 1, higher preoperative exoPD-L1 or sPD-L1 significantly impaired RFS (exoPD-L1, P = 0.0043; sPD-L1, P = 0.0041) and OS (exoPD-L1, P = 0.0034; sPD-L1, P = 0.0061). Furthermore, preoperative exoPD-L1 was negatively correlated with CD3 + T-lymphocytes infiltrated at tumor center (CT), and GB and PD1 were expressed at tumor invasive margin (IM). Preoperative sPD-L1 was negatively correlated with CD3 + and CD8 + T-lymphocytes' infiltration at IM and CT, GB and PD1 expression at IM. In cohort 2, exoPD-L1 and sPD-L1 levels decreased following hepatectomy but increased when tumor progressed. Moreover, higher postoperative exoPD-L1 and sPD-L1 or a small reduction in exoPD-L1 and sPD-L1 levels after hepatectomy suggested higher early recurrence rate. CONCLUSIONS: Both preoperative exoPD-L1 and sPD-L1 had promising prognostic values and were associated with T cell infiltration at liver metastases in CRLM patients following hepatectomy. Dynamically tracking exoPD-L1 and sPD-L1 levels could monitor disease status and detect early recurrence.


Subject(s)
B7-H1 Antigen/blood , Biomarkers, Tumor , Colorectal Neoplasms/pathology , Liver Neoplasms/blood , Liver Neoplasms/secondary , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Adult , Aged , Aged, 80 and over , B7-H1 Antigen/genetics , Cell-Derived Microparticles/metabolism , Exosomes/metabolism , Female , Gene Expression , Hepatectomy , Humans , Immunohistochemistry , Immunomodulation , Kaplan-Meier Estimate , Liquid Biopsy , Liver Neoplasms/mortality , Liver Neoplasms/surgery , Lymphocytes, Tumor-Infiltrating/immunology , Male , Middle Aged , Postoperative Period , Prognosis , Recurrence , Young Adult
17.
Rheumatology (Oxford) ; 61(2): 815-825, 2022 02 02.
Article in English | MEDLINE | ID: mdl-33930105

ABSTRACT

OBJECTIVE: To investigate the role of programmed cell death protein 1 (PD-1) and its two ligands, PD-L1 and PD-L2, in the pathogenesis of IgG4-related disease (IgG4-RD). METHODS: Patients with IgG4-RD (n = 43) and healthy controls (n = 34) were recruited. Expression levels of PD-1, PD-L1 and PD-L2 in plasma, submandibular gland and T cell subsets were determined by ELISA, immunohistochemistry and flow cytometry. Naïve T cells were stimulated with or without PD-L1/PD-L2 or anti-PD-L1/anti-PD-L2 for 7 days and the proportion of CD4+CD25+ Treg cells was detected by flow cytometry. RESULTS: The expression of PD-1, PD-L1 and PD-L2 in the plasma, submandibular gland and on the surface of Treg cells was increased in IgG4-RD patients. Plasma soluble (s)PD-1 was positively correlated with serum IgG, IgG1, IgG3, IgG4, IgG4-RD responder index and numbers of organs involved, and negatively correlated with serum IgM, IgA, C3 and C4. Plasma sPD-L2 was positively correlated with serum IgG1, and plasma sPD-L1 was positively correlated with sPD-L2 and negatively correlated with C3. Stimulation of PD-L1 but not PD-L2 promoted the differentiation of naïve T cells from IgG4-RD patients into CD4+CD25+ Treg cells. CONCLUSION: Plasma concentrations of sPD-1, sPD-L1 and sPD-L2 were significantly increased in patients with IgG4-RD, and the expression of PD-1 and PD-L2 on Treg cells was upregulated. PD-1-PD-L1 can promote the differentiation of naïve T cells into Treg cells and thus participate in the pathogenesis of IgG4-RD.


Subject(s)
B7-H1 Antigen/metabolism , Immunoglobulin G4-Related Disease/etiology , Programmed Cell Death 1 Ligand 2 Protein/metabolism , Programmed Cell Death 1 Receptor/metabolism , B7-H1 Antigen/blood , CD4-Positive T-Lymphocytes/metabolism , Case-Control Studies , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Humans , Immunoglobulin G4-Related Disease/blood , Immunoglobulin G4-Related Disease/metabolism , Programmed Cell Death 1 Ligand 2 Protein/blood , Programmed Cell Death 1 Receptor/blood , Submandibular Gland/metabolism , T-Lymphocytes, Regulatory/metabolism
18.
Future Oncol ; 18(2): 261-273, 2022 Jan.
Article in English | MEDLINE | ID: mdl-34874185

ABSTRACT

Background: We performed a meta-analysis to evaluate the association between soluble PD-L1 (sPD-L1) and survival outcomes and treatment response in lung cancer. Methods & methods: Eligible studies were obtained by searching PubMed, EMBASE and Web of Science. Pooled effect estimates were calculated for overall survival (OS), progression-free survival (PFS) and objective response rate (ORR). Results: Twelve eligible studies with 1188 lung cancer patients were included. High sPD-L1 was significantly associated with worse OS (hazard ratio [HR] = 2.20; 95% CI: 1.59-3.05; p < 0.001) and PFS (HR = 2.42; 95% CI: 1.72-3.42; p < 0.001) in patients treated with immune checkpoint inhibitors (ICIs). Meanwhile, high sPD-L1 predicted worse OS (HR = 1.60; 95% CI: 1.31-1.96; p < 0.001) and lower ORR (odds ratio = 0.52; 95% CI: 0.35-0.80; p = 0.002) in patients treated with non-ICI therapies. Conclusion: sPD-L1 is a potential predictive biomarker of lung cancer.


Lay abstract PD-L1 is a molecule that may suppress immune response to tumor by binding to its receptor PD-1. Agents blocking PD-L1/PD-1 pathway have greatly improved survival in many cancer patients. However, biomarkers that help select patients who can respond to these agents versus those who cannot are important. The soluble form of PD-L1 (sPD-L1) in peripheral blood (sPD-L1) can be easily detected. By pooling available evidence via meta-analysis, we find that lung cancer patients with high sPD-L1 level are less likely to response to anti-PD-L1/PD-1 antibodies, may progress earlier and have a shorter survival time than those with lower sPD-L1 level after treatment. Thus, sPD-L1 can be used as a biomarker predicting treatment response and survival in lung cancer. Yet some issues ­ for example, the cutoff and standard detection of sPD-L1 ­ need to be resolved for its clinical utility.


Subject(s)
B7-H1 Antigen/blood , Biomarkers, Tumor/blood , Carcinoma, Non-Small-Cell Lung/mortality , Immune Checkpoint Inhibitors/therapeutic use , Lung Neoplasms/mortality , B7-H1 Antigen/antagonists & inhibitors , B7-H1 Antigen/metabolism , Biomarkers, Tumor/antagonists & inhibitors , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/blood , Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , Lung Neoplasms/blood , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Prognosis , Progression-Free Survival
19.
JCI Insight ; 7(1)2022 01 11.
Article in English | MEDLINE | ID: mdl-34874919

ABSTRACT

Immune checkpoint therapy targeting the PD-1/PD-L1 axis is a potentially novel development in anticancer therapy and has been applied to clinical medicine. However, there are still some problems, including a relatively low response rate, innate mechanisms of resistance against immune checkpoint blockades, and the absence of reliable biomarkers to predict responsiveness. In this study of in vitro and in vivo models, we demonstrate that PD-L1-vInt4, a splicing variant of PD-L1, plays a role as a decoy in anti-PD-L1 antibody treatment. First, we showed that PD-L1-vInt4 was detectable in clinical samples and that it was possible to visualize the secreting variants with IHC. By overexpressing the PD-L1-secreted splicing variant on MC38 cells, we observed that an immune-suppressing effect was not induced by their secretion alone. We then demonstrated that PD-L1-vInt4 secretion resisted anti-PD-L1 antibody treatment, compared with WT PD-L1, which was explicable by the PD-L1-vInt4's decoying of the anti-PD-L1 antibody. The decoying function of PD-L1 splicing variants may be one of the reasons for cancers being resistant to anti-PD-L1 therapy. Measuring serum PD-L1 levels might be helpful in deciding the therapeutic strategy.


Subject(s)
B7-H1 Antigen , Drug Resistance, Neoplasm/genetics , Immune Checkpoint Inhibitors , Immunotherapy , Lung Neoplasms , Animals , B7-H1 Antigen/blood , B7-H1 Antigen/genetics , B7-H1 Antigen/metabolism , Cell Line, Tumor , Female , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mice , Mice, Inbred C57BL , Polyadenylation/genetics , Protein Isoforms/blood , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism
20.
Am J Respir Crit Care Med ; 205(1): 46-59, 2022 01 01.
Article in English | MEDLINE | ID: mdl-34731593

ABSTRACT

Rationale: Sepsis is the leading cause of death in adult ICUs. At present, sepsis diagnosis relies on nonspecific clinical features. It could transform clinical care to have immune-cell biomarkers that could predict sepsis diagnosis and guide treatment. For decades, neutrophil phenotypes have been studied in sepsis, but a diagnostic cell subset has yet to be identified. Objectives: To identify an early, specific immune signature of sepsis severity that does not overlap with other inflammatory biomarkers and that distinguishes patients with sepsis from those with noninfectious inflammatory syndrome. Methods: Mass cytometry combined with computational high-dimensional data analysis was used to measure 42 markers on whole-blood immune cells from patients with sepsis and control subjects and to automatically and comprehensively characterize circulating immune cells, which enables identification of novel, disease-specific cellular signatures. Measurements and Main Results: Unsupervised analysis of high-dimensional mass cytometry data characterized previously unappreciated heterogeneity within the CD64+ immature neutrophils and revealed two new subsets distinguished by CD123 and PD-L1 (programmed death ligand 1) expression. These immature neutrophils exhibited diminished activation and phagocytosis functions. The proportion of CD123-expressing neutrophils correlated with clinical severity. Conclusions: This study showed that these two new neutrophil subsets were specific to sepsis and detectable through routine flow cytometry by using seven markers. The demonstration here that a simple blood test distinguishes sepsis from other inflammatory conditions represents a key biological milestone that can be immediately translated into improvements in patient care.


Subject(s)
B7-H1 Antigen/blood , Interleukin-3 Receptor alpha Subunit/blood , Neutrophils/metabolism , Sepsis/diagnosis , Biomarkers/blood , Case-Control Studies , Clinical Decision Rules , Diagnosis, Differential , Flow Cytometry , Humans , Linear Models , Longitudinal Studies , Receptors, IgG/blood , Sensitivity and Specificity , Sepsis/blood , Sepsis/immunology , Severity of Illness Index
SELECTION OF CITATIONS
SEARCH DETAIL
...