Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
J Clin Oncol ; 40(6): 681-692, 2022 02 20.
Article in English | MEDLINE | ID: mdl-34985934

ABSTRACT

Malignant pleural mesothelioma (MPM) is a rare malignancy with few treatment options. Recent advances have led to US Food and Drug Administration approvals and changes in the standard of care with a novel biomedical device approved for use with platinum-pemetrexed, and also for immunotherapy agents to be included as a frontline treatment option in unresectable disease. Although predictive biomarkers for systemic therapy are not currently in use in clinical practice, it is essential to correctly identify the MPM histology to determine an optimal treatment plan. Patients with nonepithelioid MPM may have a greater magnitude of benefit to dual immunotherapy checkpoint inhibitors and this regimen should be preferred in the frontline setting for these patients. However, all patients with MPM can derive benefit from immunotherapy treatments, and these agents should ultimately be used at some point during their treatment journey. There are ongoing studies in the frontline unresectable setting that may further define the frontline therapy space, but a critical area of research will need to focus on the immunotherapy refractory population. This review article will describe the new developments in the areas of biology with genomics and chromothripsis, and also focus on updates in treatment strategies in radiology, surgery, radiation, and medical oncology with cellular therapies. These recent innovations are generating momentum to find better therapies for this disease.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy , Mesothelioma, Malignant/therapy , Pleural Neoplasms/therapy , Radiotherapy, Intensity-Modulated , Thoracic Surgical Procedures , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Cell- and Tissue-Based Therapy , Chemotherapy, Adjuvant , Humans , Immunotherapy/adverse effects , Immunotherapy/mortality , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/mortality , Mesothelioma, Malignant/pathology , Pleural Neoplasms/immunology , Pleural Neoplasms/mortality , Pleural Neoplasms/pathology , Radiation Dosage , Radiotherapy, Adjuvant , Radiotherapy, Intensity-Modulated/adverse effects , Radiotherapy, Intensity-Modulated/mortality , Thoracic Surgical Procedures/adverse effects , Thoracic Surgical Procedures/mortality , Treatment Outcome
2.
BMC Pulm Med ; 21(1): 373, 2021 Nov 15.
Article in English | MEDLINE | ID: mdl-34781910

ABSTRACT

BACKGROUND: Nivolumab is used for the treatment of malignant pleural mesothelioma (MPM). However, immune-related adverse events (irAEs) occur in patients treated with nivolumab. Several studies have reported the correlation between irAEs and therapeutic effects of immune checkpoint inhibitor, but none have reported the correlation in MPM. Here we report a retrospective study which shows the correlation between irAEs and therapeutic effects of nivolumab in patients with MPM. METHODS: This study included patients treated with nivolumab at Tokushima University Hospital from February 2009 to September 2021. We retrospectively reviewed the medical records to evaluate the several clinical factors, such as the presence or absence of irAEs, their severities, progression-free survival (PFS), overall survival (OS) or objective response to the treatment. RESULTS: Eleven patients received treatment with nivolumab. Objective response rate was 18.2% and the disease control rate was 90.9%. Median PFS was 6.8 months (95% confidence interval, 1.3 to 11.9 months) and median OS was 15.2 months (95% confidence interval, 8.9 to 21.5 months). IrAEs occurred in eight patients (72.7%), and grade ≥ 2 irAEs occurred in six patients (54.5%). PFS and OS were significantly longer in the grade ≥ 2 irAEs group than in grade < 2 irAEs group (median PFS 13.6 vs. 3.8 months, p = 0.0093; median OS not reached vs. 8.6 months, p = 0.0108). CONCLUSIONS: This is the first study to report the correlation between irAEs and therapeutic effects in patients with MPM. Because the presence of irAEs may be associated with a favorable clinical outcome, early detection and appropriate management of irAEs will increase the therapeutic benefits to patients.


Subject(s)
Antineoplastic Agents, Immunological/adverse effects , Immune System Diseases/chemically induced , Mesothelioma, Malignant/drug therapy , Nivolumab/adverse effects , Aged , Aged, 80 and over , Antineoplastic Agents, Immunological/immunology , Antineoplastic Agents, Immunological/therapeutic use , Female , Humans , Male , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/mortality , Middle Aged , Nivolumab/immunology , Nivolumab/therapeutic use , Retrospective Studies , Survival Analysis , Treatment Outcome
3.
Anticancer Res ; 41(10): 4837-4855, 2021 Oct.
Article in English | MEDLINE | ID: mdl-34593432

ABSTRACT

BACKGROUND/AIM: The adenovirus vector- carrying reduced expression in immortalized cell (REIC) gene (Ad-REIC) increases endoplasmic reticulum stress chaperone GRP78/BiP expression and induces the JNK-mediated apoptotic pathway. We aimed to determine whether Ad-REIC-induced apoptotic cell death can trigger immunogenic cell death (ICD). MATERIALS AND METHODS: We examined the emission of damage-associated molecular patterns in vitro and the vaccination effect in vivo. We determined the immunological changes in the tumour microenvironment by putative ICD inducers and the combined effects of immune checkpoint blockade therapies. RESULTS: Ad-REIC induced the release of high-mobility group box 1 and adenosine triphosphate and the translocation of calreticulin in murine mesothelioma AB12 cells. The vaccination effect was elicited by Ad-REIC treatment in vivo. The effect of Ad-REIC was potentiated by anti-cytotoxic T-lymphocyte-associated protein 4 antibody treatment in a murine mesothelioma AB1-HA cell model. CONCLUSION: Ad-REIC induces ICD in malignant mesothelioma.


Subject(s)
Adaptor Proteins, Signal Transducing/administration & dosage , Cancer Vaccines/administration & dosage , Immunogenic Cell Death/drug effects , Mesothelioma, Malignant/therapy , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/immunology , Adenosine Triphosphate/metabolism , Adenoviridae/genetics , Animals , Apoptosis/drug effects , CD8 Antigens/metabolism , Calreticulin/metabolism , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Combined Modality Therapy , Endoplasmic Reticulum Chaperone BiP , Genetic Therapy , Genetic Vectors , HMGB1 Protein/metabolism , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Inhibitors/therapeutic use , Mesothelioma, Malignant/immunology , Mice , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology , Xenograft Model Antitumor Assays
4.
Sci Rep ; 11(1): 19138, 2021 09 27.
Article in English | MEDLINE | ID: mdl-34580349

ABSTRACT

Pleural mesothelioma is an aggressive malignancy with limited effective therapies. In order to identify therapeutic targets, we integrated SNP genotyping, sequencing and transcriptomics from tumours and low-passage patient-derived cells. Previously unrecognised deletions of SUFU locus (10q24.32), observed in 21% of 118 tumours, resulted in disordered expression of transcripts from Hedgehog pathways and the T-cell synapse including VISTA. Co-deletion of Interferon Type I genes and CDKN2A was present in half of tumours and was a predictor of poor survival. We also found previously unrecognised deletions in RB1 in 26% of cases and show sub-micromolar responses to downstream PLK1, CHEK1 and Aurora Kinase inhibitors in primary mesothelioma cells. Defects in Hippo pathways that included RASSF7 amplification and NF2 or LATS1/2 mutations were present in 50% of tumours and were accompanied by micromolar responses to the YAP1 inhibitor Verteporfin. Our results suggest new therapeutic avenues in mesothelioma and indicate targets and biomarkers for immunotherapy.


Subject(s)
Biomarkers, Tumor/genetics , Gene Expression Regulation, Neoplastic/immunology , Hippo Signaling Pathway/genetics , Mesothelioma, Malignant/genetics , Pleural Neoplasms/genetics , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/antagonists & inhibitors , Biopsy , DNA Copy Number Variations , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Genomics , Hippo Signaling Pathway/drug effects , Hippo Signaling Pathway/immunology , Humans , Male , Mesothelioma, Malignant/drug therapy , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/pathology , Middle Aged , Mutation , Pleura/pathology , Pleural Neoplasms/drug therapy , Pleural Neoplasms/immunology , Pleural Neoplasms/pathology , Primary Cell Culture , Whole Genome Sequencing
5.
Eur J Cancer ; 152: 60-67, 2021 07.
Article in English | MEDLINE | ID: mdl-34087572

ABSTRACT

INTRODUCTION: Recent clinical trials with immune checkpoint inhibitors (ICIs) have shown that a subgroup of patients with malignant pleural mesothelioma (MPM) could benefit from these agents. However, there are no accurate biomarkers to predict who will respond. The aim of this study was to assess the accuracy of exhaled breath analysis using electronic technology (eNose) for discriminating between responders to ICI and non-responders. METHODS: This proof-of-concept prospective observational study was part of an intervention study (INITIATE) in patients with recurrent MPM who were treated with nivolumab (anti-PD-1) plus ipilimumab (anti-CTLA-4). At baseline and after six weeks of treatment, breath profiles were collected by an eNose. Modified Response Evaluation Criteria in Solid Tumors were used to assess efficacy at 6-month follow-up. For data processing and statistics, we used independent t-test analyses followed by linear discriminant and receiver-operating characteristic (ROC) analysis. RESULTS: Exhaled breath data of 31 MPM patients who received nivolumab plus ipilimumab were available at baseline. There were 16 with and 15 without a response after 6 months of treatment. At baseline, breath profiles significantly differed between responders and non-responders, with a cross validation value of 71%. The ROC-AUC after internal cross-validation was 0.90 (confidence interval: 0.80-1.00). CONCLUSION: An eNose is able to discriminate at baseline between responders and non-responders to nivolumab plus ipilimumab in MPM, thereby potentially identifying a subgroup of patients that will benefit from ICI treatment.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Immune Checkpoint Inhibitors/administration & dosage , Mesothelioma, Malignant/drug therapy , Neoplasm Recurrence, Local/drug therapy , Pleural Neoplasms/drug therapy , Adult , Aged , Breath Tests/instrumentation , Female , Follow-Up Studies , Humans , Male , Mesothelioma, Malignant/immunology , Middle Aged , Neoplasm Recurrence, Local/immunology , Pleural Neoplasms/immunology , Prognosis , Prospective Studies , Response Evaluation Criteria in Solid Tumors , Treatment Outcome
6.
Cytokine ; 146: 155622, 2021 10.
Article in English | MEDLINE | ID: mdl-34153874

ABSTRACT

BACKGROUND: No data on circulating biomarkers for the prognostic stratification of Malignant Pleural Mesothelioma (MPM) patients are available. We prospectively explored the prognostic role of circulating monocyte and cytokine levels and their dynamic change during chemotherapy. PATIENTS AND METHODS: MPM patients receiving a first line treatment based on a platinum compound plus pemetrexed were eligible. Blood samples were collected at the baseline and at the end of induction chemotherapy. CCL-2, IL-10 and TGF-ß levels in plasma were quantified by Enzyme-Linked Immunosorbent Assay (ELISA); white blood cells, monocytes and platelets were evaluated by blood count test. RESULTS: Thirty-one patients were included in the study. Median overall survival (OS) was 12.13 months versus 9.6 months in patients with lower and higher monocytes count, respectively (p value = 0.02). We further stratified patients according to a combined score based on the association of IL-10, TGF-ß levels and monocytes count. High combined score was associated with shorter OS and PFS in univariate and multivariate analysis. Chemotherapy induced an increase in monocytes, IL-10, but not TGF-ß levels. CONCLUSION: The prognostic value of circulating levels of multiple immunosuppressive cytokines and inflammatory cells should be confirmed in a wider validation set of MPM patients.


Subject(s)
Cytokines/blood , Cytokines/immunology , Immunosuppression Therapy , Mesothelioma, Malignant/blood , Mesothelioma, Malignant/immunology , Pleural Neoplasms/blood , Pleural Neoplasms/immunology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/blood , Humans , Kaplan-Meier Estimate , Mesothelioma, Malignant/drug therapy , Middle Aged , Monocytes/metabolism , Pleural Neoplasms/drug therapy , Prognosis , Treatment Outcome
7.
Expert Opin Emerg Drugs ; 26(2): 179-192, 2021 06.
Article in English | MEDLINE | ID: mdl-33945357

ABSTRACT

Introduction: Malignant pleural mesothelioma (MPM) has limited treatment options with minimal new therapy approvals for unresectable disease in the past 15 years. However, considerable work has occurred to develop immunotherapies and biomarker driven therapy to improve patient outcomes over this period.Areas covered: This review examines current standard of care systemic therapy in the first- and second line setting. The last 12 months has seen 2 significant trials (Checkmate 743 and CONFIRM) which provide evidence supporting the role of immunotherapy in the management of MPM. Further trials are underway to assess the role of combination chemoimmunotherapy and personalized therapy. Additionally, a large number of clinical trials are ongoing to assess the efficacy of oncoviral, dendritic cell, anti-mesothelin and chimeric antigen receptor T cell therapy in the treatment of MPM.Expert opinion: Recent Phase III trial results have established a role for immunotherapy in the management of MPM. The optimal sequencing and combination of chemotherapy and immunotherapy remains to be determined. Novel therapies for MPM are promising however efficacy remains to be determined and issues remain regarding access to and delivery of these therapies.


Subject(s)
Immunotherapy/methods , Mesothelioma, Malignant/therapy , Pleural Neoplasms/therapy , Biological Therapy/methods , Biomarkers, Tumor/metabolism , Humans , Mesothelioma, Malignant/immunology , Pleural Neoplasms/immunology , Precision Medicine
8.
Cancer Immunol Immunother ; 70(11): 3349-3355, 2021 Nov.
Article in English | MEDLINE | ID: mdl-34003301

ABSTRACT

BACKGOUND: Literature reports suggest that the host immune system may control Malignant Pleural Mesothelioma (MPM) growth, although its activity is limited by regulatory mechanisms. In this retrospective study, we analyzed the levels of pro-inflammatory (IL-1, IL-6, TNF), immune-regulatory (IL-10) and Th1/CTL-related cytokines (IL-12p70, IFN-γ) in the pleural exudate and their relationship with overall survival (OS) in MPM. METHODS: Cytokines were quantified by multiplexed immunoassay. Concentrations were dichotomized with respect to the median value. Correlation between cytokine level and OS was assessed using univariate (Kaplan-Meier curves) and multivariate (Cox regression) analyses. RESULTS: Regarding outcome, tumor histology, therapies undergone and IFN-γ were independent prognostic factors of OS in a 72 MPM training cohort. Notably, high concentrations of IFN-γ halved death probability (HR of high vs low IFN-γ concentration = 0.491, 95%CI 0.3-0.8, p = 0.007). Also in patients with epithelioid histology and those receiving at least one line of therapy, high IFN-γ level was an independent factor predictive of OS (HR of high vs low IFN-γ concentration were 0.497, p = 0.007 and 0.324, p = 0.006, respectively). However, these data were not confirmed in a 77 MPM validation cohort, possibly due to the low IFN-γ levels encountered in this population, and the heterogeneous distribution of disease stages between the training and the validation cohorts. None of the other cytokines showed any effect on survival. CONCLUSIONS: High level of IFN-γ in pleural effusion may be associated with better survival in MPM patients and potentially serve as a prognostic biomarker. Larger prospective studies are needed to ascertain this hypothesis.


Subject(s)
Interferon-gamma/metabolism , Mesothelioma, Malignant/pathology , Pleural Effusion, Malignant/metabolism , Pleural Neoplasms/pathology , Adult , Aged , Cytokines/analysis , Female , Humans , Male , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/mortality , Middle Aged , Pleural Effusion, Malignant/immunology , Pleural Neoplasms/immunology , Pleural Neoplasms/mortality , Prognosis , Retrospective Studies
9.
Virchows Arch ; 479(5): 1047-1050, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33721119

ABSTRACT

The separation of benign from malignant mesothelial cells is often a challenging problem. Some studies have suggested that immunohistochemical staining of CD146 can be used to make this distinction, but there are marked differences in the reported results. Here, we assessed CD146 expression in tissue microarray specimens of 32 epithelioid reactive mesothelial hyperplasias, 17 spindle cell reactive mesothelial proliferations, 43 epithelioid mesotheliomas, and 31 sarcomatoid mesotheliomas. We found that, although the specificity of CD146 for epithelioid mesotheliomas versus reactive epithelial mesothelial proliferations was high (94%), staining intensity and extent was usually low and sensitivity was poor (23%). For sarcomatoid mesotheliomas versus reactive spindle cell mesothelial processes, both measures (33% sensitivity, 76% specificity) were inadequate. Furthermore, strong staining of endothelial cells and fibroblasts often created difficulties in interpretation. In comparison, BAP1 was lost in 21/43 (49%) epithelioid and 9/31 (29%) sarcomatoid mesotheliomas and methylthioadenosine phosphorylase (MTAP) was lost in 9/40 (23%) epithelioid and 7/29 (24%) sarcomatoid mesotheliomas from these TMAs. There was no association between CD146 staining and BAP1 or MTAP retention/loss. We conclude that CD146 staining is probably not useful for separating malignant from benign mesothelial proliferations.


Subject(s)
Biomarkers, Tumor/analysis , Cell Proliferation , Immunohistochemistry , Mesothelioma, Malignant/immunology , CD146 Antigen/analysis , Diagnosis, Differential , Humans , Mesothelioma, Malignant/pathology , Predictive Value of Tests , Tissue Array Analysis
10.
Int J Oncol ; 58(4)2021 04.
Article in English | MEDLINE | ID: mdl-33655329

ABSTRACT

Exposure of human immune cells to asbestos causes a reduction in antitumor immunity. The present study aimed to investigate the recovery of reduced antitumor immunity by several ingredients taken as supplements or foods, including trehalose (Treh) and glycosylated hesperidin (gHesp). Peripheral blood CD4+ cells were stimulated with IL­2, anti­CD3 and anti­CD28 antibodies for 3 days, followed by further stimulation with IL­2 for 7 days. Subsequently, cells were stimulated with IL­2 for an additional 28 days. During the 28 days, cells were cultured in the absence or presence of 50 µg/ml chrysotile asbestos fibers. In addition, cells were treated with 10 mM Treh or 10 µM gHesp. Following culture for 28 days, reverse transcription­quantitative PCR was performed to assess the expression levels of transcription factors, cytokines and specific genes, including matrix metalloproteinase­7 (MMP­7), nicotinamide nucleotide transhydrogenase (NNT) and C­X­C motif chemokine receptor 3, in unstimulated cells (fresh) and cells stimulated with PMA and ionomycin (stimuli). The results demonstrated that compared with the control group, chrysotile­exposure induced alterations in MMP­7, NNT and IL­17A expression levels were not observed in the 'Treh' and 'gHesp' groups in stimulated cells. The results suggested that Treh and gHesp may reverse asbestos exposure­induced reduced antitumor immunity in T helper cells. However, further investigation is required to confirm the efficacy of future trials involving the use of these compounds with high­risk human populations exposed to asbestos, such as workers involved in asbestos­handling activities.


Subject(s)
Asbestos/adverse effects , CD4-Positive T-Lymphocytes/immunology , Dietary Supplements , Hesperidin/pharmacology , Mesothelioma, Malignant/immunology , Trehalose/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cells, Cultured , Humans , Interferon-gamma/immunology , Interleukin-17/immunology , Male , Mesothelioma, Malignant/chemically induced , Mesothelioma, Malignant/prevention & control , Middle Aged , Receptors, CXCR3/immunology
11.
J Toxicol Environ Health A ; 84(11): 475-483, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33678145

ABSTRACT

Asbestos exposure is associated with many adverse health conditions including malignant mesothelioma and lung cancer as well as production of autoantibodies. Autoantibodies may serve as biomarkers for asbestos exposure in patients with cancer, and autoimmune dysfunction has been linked to increased rates of various cancers. The aim of this study was to examine the hypothesis that autoantibodies are more frequent in asbestos-exposed individuals with either lung cancer or mesothelioma than those without these conditions. Asbestos-exposed individuals from Western Australia who had lung cancer (n = 24), malignant mesothelioma (n = 24), or no malignancy (n = 51) were tested for antinuclear autoantibodies (ANA) using indirect immunofluorescence and specific extractable nuclear autoantibodies (ENA) employing a multiplexed addressable laser bead immunoassay. Contrary to the hypothesis, data demonstrated that individuals without malignancy were more likely to be positive for ANA compared to those with cancer. However, autoantibodies to histone and Ro-60 were found to be associated with lung cancer. These results support a possible predictive value for specific autoantibodies in the early detection of lung cancer and/or in our understanding of the role of autoimmune processes in cancer. However, further studies are needed to identify specific target antigens for the antibodies.


Subject(s)
Asbestos/adverse effects , Autoantibodies/blood , Lung Neoplasms/immunology , Mesothelioma, Malignant/immunology , Occupational Exposure/adverse effects , Aged , Female , Humans , Lung Neoplasms/chemically induced , Male , Mesothelioma, Malignant/chemically induced , Middle Aged , Mining , Western Australia
12.
J Immunother Cancer ; 9(2)2021 02.
Article in English | MEDLINE | ID: mdl-33632900

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) is a rare and aggressive neoplasia affecting the lung mesothelium. Immune checkpoint inhibitors (ICI) in MPM have not been extremely successful, likely due to poor identification of suitable candidate patients for the therapy. We aimed to identify cellular immune fractions associated with clinical outcome and classify patients with MPM based on their immune contexture. For each defined group, we sought for molecular specificities that could help further define our MPM classification at the genomic and transcriptomic level, as well as identify differential therapeutic strategies based on transcriptional signatures predictive of drug response. METHODS: The abundance of 20 immune cell fractions in 516 MPM samples from 7 gene expression datasets was inferred using gene set variation analysis. Identification of clinically relevant fractions was performed with Cox proportional-hazards models adjusted for age, stage, sex, and tumor histology. Immune-based groups were defined based on the identified fractions. RESULTS: T-helper 2 (TH2) and cytotoxic T (TC) cells were found to be consistently associated with overall survival. Three immune clusters (IG) were subsequently defined based on TH2 and TC immune infiltration levels: IG1 (54.5%) was characterized by high TH2 and low TC levels, IG2 (37%) had either low or high levels of both fractions, and IG3 (8.5%) was defined by low TH2 and high TC levels. IG1 and IG3 groups were associated with worse and better overall survival, respectively. While no differential genomic alterations were identified among immune groups, at the transcriptional level, IG1 samples showed upregulation of proliferation signatures, while IG3 samples presented upregulation of immune and inflammation-related pathways. Finally, the integration of gene expression with functional signatures of drug response showed that IG3 patients might be more likely to respond to ICI. CONCLUSIONS: This study identifies a novel immune-based signature with potential clinical relevance based on TH2 and TC levels, unveiling a fraction of patients with MPM with better prognosis and who might benefit from immune-based therapies. Molecular specificities of the different groups might be used to tailor specific potential therapies in the future.


Subject(s)
Gene Expression Profiling/methods , Gene Regulatory Networks , Mesothelioma, Malignant/genetics , T-Lymphocytes, Cytotoxic/immunology , Th2 Cells/immunology , Adult , Aged , Aged, 80 and over , Databases, Genetic , Female , Gene Expression Regulation, Neoplastic , Humans , Male , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/pathology , Middle Aged , Survival Analysis , Tumor Microenvironment , Young Adult
13.
J Cancer Res Clin Oncol ; 147(2): 459-468, 2021 Feb.
Article in English | MEDLINE | ID: mdl-33216211

ABSTRACT

PURPOSE: Programmed death-ligand 1 (PD-L1) protein plays a central role in the antitumor immune response, and appears to be a predictor of prognosis and efficacy for PD-L1 and programmed death 1 (PD-1) blockade therapy. The immunoregulatory role and prognostic impact of PD-L1 soluble form (sPD-L1) have been investigated in biological fluids of patients with different tumors. In malignant pleural mesothelioma (MPM), circulating sPD-L1 has been recently reported in patients' sera, but no data are available in pleural effusions (PE). In our study, we evaluated the baseline expression levels of sPD-L1 in PE from 84 MPM patients and correlated them with PD-L1-status in matched tumors and patients' overall survival (OS). METHODS: sPD-L1 in PE was determined by ELISA and tumor PD-L1 by immunohistochemistry. Association of sPD-L1 with OS was estimated using the Cox regression model. RESULTS: We observed that sPD-L1 was variably expressed in all the PE and tended to be higher (by 30%) in patients with PD-L1-positive tumors (cut-off ≥ 1% stained cells) as compared to patients with PD-L1-negative tumors (geometric mean ratio = 1.28, P value = 0.288). sPD-L1 levels were significantly higher than those of sPD-1 (P value = 0.001) regardless of the MPM histotypes and they were positively correlated (r = 0.50, P value < 0.001). Moreover, high PE sPD-L1 concentrations were associated with a trend towards increased OS (hazard ratio 0.79, 95% CL 0.62-1.01, P value = 0.062). CONCLUSIONS: Our study documents the presence of sPD-L1 in PE of MPM patients, and suggests its possible biological and prognostic role in MPM.


Subject(s)
B7-H1 Antigen/physiology , Mesothelioma, Malignant/immunology , Pleural Effusion/metabolism , Aged , Aged, 80 and over , B7-H1 Antigen/analysis , Female , Humans , Immunohistochemistry , Male , Mesothelioma, Malignant/mortality , Middle Aged , Prognosis , Programmed Cell Death 1 Receptor/analysis , Proportional Hazards Models
14.
Pathol Res Pract ; 216(12): 153277, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33223279

ABSTRACT

Previous studies have reported a close relationship between type V collagen (Col V) and tumor invasion and motility in both breast cancer (BC) and lung cancer (LC). The present work aims to determine whether the extracellular-matrix (ECM)-defined microenvironment influences patient clinical outcome and investigate to which extent histological patterns of Col V expression in malignant cells have a prognostic effect in patients. To that end, we examined the expression of Col V in the tissues of 174 primary tumors (MM, N = 82; LC, N = 41; and BC, N = 46) by immunohistochemistry. We found: (1) diffuse strong green birefringence in membrane and cytoplasm individualizing malignant cells in MM; (2) a focal and weak birefringence mainly in cytoplasmic membrane involving groups of malignant cells in LC and BC; (3) higher average H-score of Col V in MM than in LC and BC samples; (4) a direct correlation between Col V histologic pattern and TNM stage IV, status and median overall survival; (5) patients with LC in TNM stage I, and Col V ≤ 41.7 IOD/mm2 had a low risk of death and a median survival time more than 20 months; (6) patients with MM in TNM stage IV and Col V > 41.7 IOD/mm2 presented a high risk of death and a median survival time of just 20 months. These findings suggest that high levels of Col V individualizing malignant cells, as observed in MM, and low levels grouping malignant cells, as observed in LC and BC, confers different immune-privileged tissue microenvironment for tumor invasion with impact on prognosis of the patients.


Subject(s)
Breast Neoplasms/chemistry , Cell Movement , Collagen Type V/analysis , Extracellular Matrix/chemistry , Lung Neoplasms/chemistry , Mesothelioma, Malignant/chemistry , Tumor Microenvironment , Aged , Breast Neoplasms/immunology , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Extracellular Matrix/pathology , Female , Humans , Immunohistochemistry , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Male , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/pathology , Middle Aged , Neoplasm Invasiveness , Neoplasm Staging , Tumor Microenvironment/immunology
15.
J Immunol ; 205(9): 2519-2531, 2020 11 01.
Article in English | MEDLINE | ID: mdl-32948683

ABSTRACT

Increasing evidence indicates that local hypofractionated radiotherapy (LRT) can elicit both immunogenic and immunosuppressive local and systemic immune responses. We thus hypothesized that blockade of LRT-induced immunosuppressive responses could augment the antitumor effects and induce an abscopal response. In this study, we found that the upregulation of Foxp3+ regulatory T cells (Tregs) in the mesothelioma tumor microenvironment after nonablative oligofractionated irradiation significantly limited the success of irradiation. Using DEREG mice, which allow conditional and efficient depletion of Foxp3+ Tregs by diphtheria toxin injection, we observed that transient Foxp3+ Treg depletion immediately after nonablative oligofractionated irradiation provided synergistic local control and biased the T cell repertoire toward central and effector memory T cells, resulting in long-term cure. Furthermore, this combination therapy showed significant abscopal effect on the nonirradiated tumors in a concomitant model of mesothelioma through systemic activation of cytotoxic T cells and enhanced production of IFN-γ and granzyme B. Although local control was preserved with one fraction of nonablative irradiation, three fractions were required to generate the abscopal effect. PD-1 and CTLA-4 were upregulated on tumor-infiltrating CD4+ and CD8+ T cells in irradiated and nonirradiated tumors, suggesting that immune checkpoint inhibitors could be beneficial after LRT and Foxp3+ Treg depletion. Our findings are applicable to the strategy of immuno-radiotherapy for generating optimal antitumor immune responses in the clinical setting. Targeting Tregs immediately after a short course of irradiation could have a major impact on the local response to irradiation and its abscopal effect.


Subject(s)
Forkhead Transcription Factors/immunology , Mesothelioma, Malignant/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , CTLA-4 Antigen/immunology , Granzymes/immunology , Immunity/immunology , Interferon-gamma/immunology , Lymphocyte Depletion/methods , Lymphocytes, Tumor-Infiltrating/immunology , Mice , Mice, Inbred BALB C , Programmed Cell Death 1 Receptor/immunology , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology
16.
Int J Mol Sci ; 21(19)2020 Sep 23.
Article in English | MEDLINE | ID: mdl-32977478

ABSTRACT

Asbestos exposure causes malignant tumors such as lung cancer and malignant mesothelioma. The effects of asbestos fibers on immunocompetent cells, however, have not been well studied. Asbestos physically comprises a fibrous substance, which differs from silica particles which are a particulate substance, although chemically it is a mineral silicate. Since silicosis patients previously exposed to silica particles often suffer from lung and autoimmune diseases, it is clear that silica exposure impairs immune tolerance. Similarly, asbestos may alter the immune system in asbestos-exposed individuals. Given that malignant tumors can result following exposure to asbestos, the attenuation of anti-tumor immunity in cases of asbestos exposure is an important area of investigation. We observed the effect of asbestos fibers on T lymphocytes, such as CD8+ cytotoxic T lymphocytes (CTLs), CD4+ helper T (Th), and regulatory T (Treg) cells, and showed that anti-tumor immunity was attenuated, as demonstrated in a system that stimulates fresh cells isolated from peripheral blood in vitro and a system that is continuously exposed to a cell line. In this manuscript, we introduce the experiments and results of studies on CTLs, as well as Th and Treg cells, and discuss how future changes in immunocompetent cells induced by asbestos fibers can be clinically linked.


Subject(s)
Asbestos/toxicity , CD8-Positive T-Lymphocytes/immunology , Immunity, Cellular , Mesothelioma, Malignant/immunology , T-Lymphocytes, Regulatory/immunology , CD8-Positive T-Lymphocytes/pathology , Humans , Mesothelioma, Malignant/chemically induced , Mesothelioma, Malignant/pathology , T-Lymphocytes, Regulatory/pathology
17.
Curr Drug Targets ; 21(15): 1606-1612, 2020.
Article in English | MEDLINE | ID: mdl-32682370

ABSTRACT

Malignant pleural mesothelioma (MPM) is a remarkably aggressive thoracic malignancy with a limited survival of only 5-12 months. However, MPM still remains unresponsive to conventional standards of treatment, including pleurectomy and decortication, extrapleural pneumonectomy for resectable disease with or without chemotherapy, and/or radiation therapy. The mechanism of carcinogenesis has not been fully elucidated, although approximately 80% of cases can still be linked to asbestos exposure. The tumor immune microenvironment (TME) has been proven to play an important role in MPM pathogenesis and treatment outcomes. Several molecular pathways have been implicated in the MPM tumor microenvironment, such as angiogenesis, apoptosis, cell cycle regulation, and stromal processes. Immunotherapy has already shown promising results in other thoracic solid tumors, such as non-small-cell lung cancer (NSCLC). However, immunotherapy has shown less convincing results in MPM than in melanoma and NSCLC. A multicenter, randomized trial (DETERMINE) proved that immune checkpoint inhibition using tremelimumab, an anti-cytotoxic T lymphocyteassociated protein 4 (CTLA-4) antibody, failed to improve median overall survival. Therefore, it is important to explore the relationship between the characteristics of the tumor microenvironment and immunotherapy. Here, we review the heterogeneity of the TME and the progress in the understanding of the immune microenvironment and immunotherapy in MPM to explore the mechanisms of resistance to immunotherapy.


Subject(s)
Immunotherapy , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/therapy , Tumor Microenvironment/immunology , Animals , B7-H1 Antigen/metabolism , Cancer-Associated Fibroblasts/immunology , Dendritic Cells/immunology , Humans , Killer Cells, Natural/immunology , Myeloid-Derived Suppressor Cells/immunology , T-Lymphocytes/immunology , Tumor-Associated Macrophages/immunology
18.
Bioconjug Chem ; 31(7): 1750-1755, 2020 07 15.
Article in English | MEDLINE | ID: mdl-32628464

ABSTRACT

Single chain antibody fragment (scFv) is a promising agent for imaging and targeted therapy. The objective of the study is to evaluate a kit formulation for 99mTc labeling of scFv for tumor imaging. The scFv was engineered to contain a cysteine tag to accommodate the specific conjugation of HYNIC and subsequent 99mTc labeling. The labeling conditions were formulated to allow instantaneous one-pot quantitative labeling. The reproducibility of labeling was evaluated at various time points during kit storage at -20 °C. In vitro cell binding experiments and HPLC analysis were performed to assess binding affinity and radiolabel stability, respectively. In vivo tumor targeting study was performed in xenograft models with biodistribution studied at 1, 3, and 24 h post-injection. The optimized kit with 5 µg SnF2, pH 5.5, and 50 µg GH along with as low as 15 µg of HYNIC-cys-scFv provided high labeling yield (>95%), high specific activity (1.8 × 107 Ci/Mol), and robust reproducibility with shelf life up to 90 days when stored at -20 °C. The in vitro cell binding study showed the labeled scFv maintained the binding capability with an apparent KD of ∼27 nM. The animal study using tumor-bearing mice showed high tumor uptake at 16.9%ID/g 24 h post-injection along with rapid blood clearance (0.18%ID/g) and kidney excretion (44%ID/g), resulting in very high contrast (tumor/muscle >200:1). A kit formulation for 99mTc labeling of scFvs targeting mesothelioma was developed based on specific HYNIC conjugation and GH (Glucoheptonate) as a coligand, producing not only high specific activity, but also improved tumor uptake. This convenient one-pot labeling method has the potential for translation into clinical use and is applicable to other scFvs as well.


Subject(s)
Isotope Labeling/methods , Mesothelioma, Malignant/therapy , Organotechnetium Compounds/chemistry , Single-Chain Antibodies/chemistry , Animals , Mesothelioma, Malignant/immunology , Mice , Organotechnetium Compounds/pharmacokinetics , Reproducibility of Results , Single-Chain Antibodies/immunology , Tissue Distribution , Xenograft Model Antitumor Assays
19.
J Immunother Cancer ; 8(1)2020 06.
Article in English | MEDLINE | ID: mdl-32581053

ABSTRACT

BACKGROUND: Malignant pleural mesothelioma (MPM) is a rare and aggressive cancer related to asbestos exposure. The tumor microenvironment content, particularly the presence of macrophages, was described as crucial for the development of the disease. This work aimed at studying the involvement of the M-CSF (CSF-1)/IL-34/CSF-1R pathway in the formation of macrophages in MPM, using samples from patients. METHODS: Pleural effusions (PEs), frozen tumors, primary MPM cells and MPM cell lines used in this study belong to biocollections associated with clinical databases. Cytokine expressions were studied using real-time PCR and ELISA. The Cancer Genome Atlas database was used to confirm our results on an independent cohort. An original three-dimensional (3D) coculture model including MPM cells, monocytes from healthy donors and a tumor antigen-specific cytotoxic CD8 T cell clone was used. RESULTS: We observed that high interleukin (IL)-34 levels in PE were significantly associated with a shorter survival of patients. In tumors, expression of CSF1 was correlated with 'M2-like macrophages' markers, whereas this was not the case with IL34 expression, suggesting two distinct modes of action of these cytokines. Expression of IL34 was higher in MPM cells compared with primary mesothelial cells. Particularly, high expression of IL34 was observed in MPM cells with an alteration of CDKN2A. Finally, using 3D coculture model, we demonstrated the direct involvement of MPM cells in the formation of immunosuppressive macrophages, through activation of the colony stimulating factor-1 receptor (CSF1-R) pathway, causing the inhibition of cytotoxicity of tumor antigen-specific CD8+ T cells. CONCLUSIONS: The M-CSF/IL-34/CSF-1R pathway seems strongly implicated in MPM and could constitute a therapeutic target to act on immunosuppression and to support immunotherapeutic strategies.


Subject(s)
Biomarkers, Tumor/metabolism , Interleukins/metabolism , Macrophage Colony-Stimulating Factor/metabolism , Mesothelioma, Malignant/pathology , Pleural Effusion/pathology , Pleural Neoplasms/pathology , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Aged , Biomarkers, Tumor/genetics , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/pathology , Cytokines/metabolism , Female , Follow-Up Studies , Humans , Interleukins/genetics , Macrophage Colony-Stimulating Factor/genetics , Macrophages/immunology , Macrophages/metabolism , Macrophages/pathology , Male , Mesothelioma, Malignant/genetics , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/metabolism , Monocytes/immunology , Monocytes/metabolism , Monocytes/pathology , Pleural Effusion/immunology , Pleural Effusion/metabolism , Pleural Neoplasms/genetics , Pleural Neoplasms/immunology , Pleural Neoplasms/metabolism , Prognosis , Receptors, Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment/immunology
20.
Cancer Med ; 9(13): 4836-4849, 2020 07.
Article in English | MEDLINE | ID: mdl-32391978

ABSTRACT

OBJECTIVE: Previous studies have reported a close relationship between malignant mesothelioma (MM) and the immune matricial microenvironment (IMM). One of the major problems in these studies is the lack of adequate adjustment for potential confounders. Therefore, the aim of this study was to identify and quantify risk factors such as IMM and various tumor characteristics and their association with the subtype of MM and survival. METHODS: We examined IMM and other tumor markers in tumor tissues from 82 patients with MM. These markers were evaluated by histochemistry, immunohistochemistry, immunofluorescence, and morphometry. Logistic regression analysis, cluster analysis, and Cox regression analysis were performed. RESULTS: Hierarchical cluster analysis revealed two clusters of MM that were independent of clinicopathologic features. The high-risk cluster included MM with high tumor cellularity, high type V collagen (Col V) fiber density, and low CD8+ T lymphocyte density in the IMM. Our results showed that the risk of death was increased for patients with MM with high tumor cellularity (OR = 1.63, 95% CI = 1.29-2.89, P = .02), overexpression of Col V (OR = 2.60, 95% CI = 0.98-6.84, P = .04), and decreased CD8 T lymphocytes (OR = 1.001, 95% CI = 0.995-1.007, P = .008). The hazard ratio for the high-risk cluster was 2.19 (95% CI = 0.54-3.03, P < .01) for mortality from MM at 40 months. CONCLUSION: Morphometric analysis of Col V, CD8+ T lymphocytes, and tumor cellularity can be used to identify patients with high risk of death from MM.


Subject(s)
Biomarkers, Tumor/analysis , Mesothelioma, Malignant/mortality , Tumor Microenvironment , CD8-Positive T-Lymphocytes , Collagen Type I/analysis , Collagen Type V/analysis , Collagen Type V/metabolism , Female , Fluorescent Antibody Technique , Humans , Immunohistochemistry , In Situ Hybridization , Lymphocyte Count , Male , Mesothelioma, Malignant/immunology , Mesothelioma, Malignant/metabolism , Mesothelioma, Malignant/pathology , Middle Aged , Regression Analysis , Retrospective Studies , Risk Assessment , Risk Factors , Tissue Array Analysis , Tumor Microenvironment/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...