Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
Add more filters

Country/Region as subject
Affiliation country
Publication year range
1.
Am J Respir Crit Care Med ; 193(9): 1023-31, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26652184

ABSTRACT

RATIONALE: We demonstrated previously that autologous tumor lysate-pulsed dendritic cell-based immunotherapy in patients with malignant pleural mesothelioma is feasible, well-tolerated, and capable of inducing immunologic responses against tumor cells. In our murine model, we found that reduction of regulatory T cells with metronomic cyclophosphamide increased the efficacy of immunotherapy. OBJECTIVES: To assess the decrease in number of peripheral blood regulatory T cells during combination therapy of low-dose cyclophosphamide and dendritic cell immunotherapy and determine the induction of immunologic responses with this treatment in patients with mesothelioma. METHODS: Ten patients with malignant pleural mesothelioma received metronomic cyclophosphamide and dendritic cell-based immunotherapy. During the treatment, peripheral blood mononuclear cells were analyzed for regulatory T cells and immunologic responses. MEASUREMENTS AND MAIN RESULTS: Administration of dendritic cells pulsed with autologous tumor lysate combined with cyclophosphamide in patients with mesothelioma was safe, the only side effect being moderate fever. Dendritic cell vaccination combined with cyclophosphamide resulted in radiographic disease control in 8 of the 10 patients. Overall survival was promising, with 7 out of 10 patients having a survival of greater than or equal to 24 months and two patients still alive after 50 and 66 months. Low-dose cyclophosphamide reduced the percentage of regulatory T cells of total CD4 cells in peripheral blood from 9.43 (range, 4.34-26.10) to 4.51 (range, 0.27-10.30) after 7 days of cyclophosphamide treatment (P = 0.02). CONCLUSIONS: Consolidation therapy with autologous tumor lysate-pulsed dendritic cell-based therapy and simultaneously reducing the tumor-induced immune suppression is well-tolerated and shows signs of clinical activity in patients with mesothelioma. Clinical trial registered with www.clinicaltrials.gov (NCT 01241682).


Subject(s)
Cyclophosphamide/therapeutic use , Dendritic Cells/immunology , Lung Neoplasms/drug therapy , Mesothelioma/drug therapy , Vaccination/methods , Adjuvants, Pharmaceutic/therapeutic use , Adult , Aged , Combined Modality Therapy/methods , Cyclophosphamide/immunology , Female , Humans , Immunosuppressive Agents/immunology , Immunosuppressive Agents/therapeutic use , Immunotherapy/methods , Lung Neoplasms/immunology , Male , Mesothelioma/immunology , Mesothelioma, Malignant , Middle Aged , Pleura , T-Lymphocytes, Regulatory/immunology , Treatment Outcome
3.
BMC Cancer ; 12: 580, 2012 Dec 05.
Article in English | MEDLINE | ID: mdl-23217146

ABSTRACT

Cancer research has devoted most of its energy over the past decades on unraveling the control mechanisms within tumor cells that govern its behavior. From this we know that the onset of cancer is the result of cumulative genetic mutations and epigenetic alterations in tumor cells leading to an unregulated cell cycle, unlimited replicative potential and the possibility for tissue invasion and metastasis. Until recently it was often thought that tumors are more or less undetected or tolerated by the patient's immune system causing the neoplastic cells to divide and spread without resistance. However, it is without any doubt that the tumor environment contains a wide variety of recruited host immune cells. These tumor infiltrating immune cells influence anti-tumor responses in opposing ways and emerges as a critical regulator of tumor growth. Here we provide a summary of the relevant immunological cell types and their complex and dynamic roles within an established tumor microenvironment. For this, we focus on both the systemic compartment as well as the local presence within the tumor microenvironment of late-stage non-small cell lung cancer (NSCLC), admitting that this multifaceted cellular composition will be different from earlier stages of the disease, between NSCLC patients. Understanding the paradoxical role that the immune system plays in cancer and increasing options for their modulation may alter the odds in favor of a more effective anti-tumor immune response. We predict that the future standard of care of lung cancer will involve patient-tailor-made combination therapies that associate (traditional) chemotherapeutic drugs and biologicals with immune modulating agents and in this way complement the therapeutic armamentarium for this disease.


Subject(s)
Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/therapy , Lung Neoplasms/immunology , Lung Neoplasms/therapy , Lymphocytes, Tumor-Infiltrating/immunology , Humans , Immunotherapy/methods , Immunotherapy/trends , Precision Medicine/methods , Precision Medicine/trends , Tumor Microenvironment/immunology
4.
BMC Pulm Med ; 12: 77, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23234250

ABSTRACT

BACKGROUND: During the past decades, numerous efforts have been made to decrease the death rate among lung cancer patients. Nonetheless, the improvement in long-term survival has been limited and lung cancer is still a devastating disease. DISCUSSION: With this article we would like to point out that survival of lung cancer could be strongly improved by controlling two pivotal prognostic factors: stage and treatment. This is corresponding with recent reports that show a decrease in lung cancer mortality by screening programs. In addition, modulation of the patient's immune system by immunotherapy either as monotherapy or combined with conventional cancer treatments offers the prospect of tailoring treatments much more precisely and has also been shown to lead to a better response to treatment and overall survival of non-small cell lung cancer patients. SUMMARY: Since only small improvements in survival can be expected in advanced disease with the use of conventional therapies, more research should be focused on lung cancer screening programs and patient tailored immunotherapy with or without conventional therapies. If these approaches are clinically combined in a standard multidisciplinary policy we might be able to advance the survival of patients with lung cancer.


Subject(s)
Early Detection of Cancer/trends , Lung Neoplasms/diagnosis , Lung Neoplasms/mortality , Drug Therapy , Humans , Immunotherapy , Lung Neoplasms/therapy , Neoplasm Staging , Prognosis , Survival Rate
5.
J Hepatol ; 54(2): 209-18, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21095036

ABSTRACT

BACKGROUND & AIMS: Natural killer (NK) cells play a major role in anti-viral immunity as first line defense and regulation of virus-specific T cell responses. This study aimed to investigate phenotype and function of NK cells in patients with chronic hepatitis B virus (HBV) infection and to study the effect of anti-viral therapy. METHODS: Peripheral blood NK cells from 40 chronic HBV patients were compared to NK cells of 25 healthy controls. The effect of entecavir-induced viral load reduction on NK cell phenotype and function was investigated in 15 chronic HBV patients. RESULTS: NK cell numbers and subset distribution did not differ between HBV patients and normal subjects. In chronic HBV patients, the cytotoxic capacity was retained, but NK cell activation and subsequent IFNγ and TNFα production, especially of the CD56(dim) subset, were strongly hampered. This functional dichotomy was paralleled by an altered activation state, elevated expression of NKG2A, and downregulated expression of CD16 and NKp30, which correlated with serum HBV-DNA load. Anti-viral therapy partially restored NK cell phenotype, as shown by NKG2A downregulation. Moreover, viral replication inhibition improved IFNγ production as a result of an increased ability of CD56(dim) NK cells to become activated de novo. This improved NK cell activation and function which correlated with therapy-induced reduction in serum ALT levels, but not HBV-DNA load. CONCLUSIONS: The specific defect in CD56(dim) NK cell activation and the reduced capacity to produce anti-viral and Th1-skewing cytokines may play a role in HBV persistence. Restoration of this NK cell cytokine-producing capacity, as achieved by viral load reduction, could therefore contribute to definite clearance of the virus.


Subject(s)
Hepatitis B, Chronic/immunology , Hepatitis B, Chronic/virology , Killer Cells, Natural/immunology , Lymphocyte Activation , Viral Load , Adult , CD56 Antigen/analysis , Cross-Sectional Studies , Cytotoxicity, Immunologic , Female , Flow Cytometry , GPI-Linked Proteins/analysis , Guanine/analogs & derivatives , Guanine/therapeutic use , HLA-DR Antigens/analysis , Hepatitis B, Chronic/drug therapy , Humans , Interferon-gamma/biosynthesis , Male , NK Cell Lectin-Like Receptor Subfamily C/analysis , Natural Cytotoxicity Triggering Receptor 3/analysis , Receptors, IgG/analysis
6.
Am J Respir Crit Care Med ; 181(12): 1383-90, 2010 Jun 15.
Article in English | MEDLINE | ID: mdl-20167848

ABSTRACT

RATIONALE: We previously demonstrated that dendritic cell-based immunotherapy induced protective antitumor immunity with a prolonged survival rate in mice. However, the clinical relevance is still in question. To examine this, we designed a clinical trial using chemotherapy followed by antigen-pulsed dendritic cell vaccination in mesothelioma patients. OBJECTIVES: The aim of this study was to assess the safety and immunological response induced by the administration of tumor lysate-pulsed dendritic cells in patients with mesothelioma. METHODS: Ten patients with malignant pleural mesothelioma received three vaccinations of clinical-grade autologous dendritic cells intradermally and intravenously at 2-week intervals after chemotherapy. Each vaccine was composed of 50 x 10(6) mature dendritic cells pulsed with autologous tumor lysate and keyhole limpet hemocyanin (KLH) as surrogate marker. Delayed-type hypersensitivity activity to tumor antigens and KLH was assessed, both in vivo and in vitro. Peripheral blood mononuclear cells during the treatment were analyzed for immunological responses. MEASUREMENTS AND MAIN RESULTS: Administration of dendritic cells pulsed with autologous tumor lysate in patients with mesothelioma was safe with moderate fever as the only side effect. There were no grade 3 or 4 toxicities associated with the vaccines or any evidence of autoimmunity. Local accumulations of infiltrating T cells were found at the site of vaccination. The vaccinations induced distinct immunological responses to KLH, both in vitro and in vivo. Importantly, after three vaccinations, cytotoxic activity against autologous tumor cells was detected in a subgroup of patients. CONCLUSIONS: This study demonstrated that autologous tumor lysate-pulsed dendritic cell-based therapy is feasible, well-tolerated, and capable of inducing immunological response to tumor cells in mesothelioma patients. Clinical trial registered with www.clinicaltrials.gov (NCT00280982).


Subject(s)
Cytotoxicity, Immunologic/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Mesothelioma/therapy , Pleural Neoplasms/therapy , Aged , Feasibility Studies , Humans , Male , Mesothelioma/immunology , Middle Aged , Pleural Neoplasms/immunology
7.
BMC Cancer ; 10: 464, 2010 Aug 30.
Article in English | MEDLINE | ID: mdl-20804550

ABSTRACT

BACKGROUND: Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature cells that accumulates in tumour-bearing hosts. These cells are induced by tumour-derived factors (e.g. prostaglandins) and have a critical role in immune suppression. MDSC suppress T and NK cell function via increased expression of arginase I and production of reactive oxygen species (ROS) and nitric oxide (NO). Immune suppression by MDSC was found to be one of the main factors for immunotherapy insufficiency. Here we investigate if the in vivo immunoregulatory function of MDSC can be reversed by inhibiting prostaglandin synthesis by specific COX-2 inhibition focussing on ROS production by MDSC subtypes. In addition, we determined if dietary celecoxib treatment leads to refinement of immunotherapeutic strategies. METHODS: MDSC numbers and function were analysed during tumour progression in a murine model for mesothelioma. Mice were inoculated with mesothelioma tumour cells and treated with cyclooxygenase-2 (COX-2) inhibitor celecoxib, either as single agent or in combination with dendritic cell-based immunotherapy. RESULTS: We found that large numbers of infiltrating MDSC co-localise with COX-2 expression in those areas where tumour growth takes place. Celecoxib reduced prostaglandin E2 levels in vitro and in vivo. Treatment of tumour-bearing mice with dietary celecoxib prevented the local and systemic expansion of all MDSC subtypes. The function of MDSC was impaired as was noticed by reduced levels of ROS and NO and reversal of T cell tolerance; resulting in refinement of immunotherapy. CONCLUSIONS: We conclude that celecoxib is a powerful tool to improve dendritic cell-based immunotherapy and is associated with a reduction in the numbers and suppressive function of MDSC. These data suggest that immunotherapy approaches benefit from simultaneously blocking cyclooxygenase-2 activity.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Immunotherapy , Mesothelioma/immunology , Mesothelioma/therapy , Myeloid Cells/immunology , Pyrazoles/pharmacology , Sulfonamides/pharmacology , Animals , Blotting, Western , Celecoxib , Cyclooxygenase 2/chemistry , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Dinoprostone/metabolism , Disease Models, Animal , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Immune Tolerance , Immunoenzyme Techniques , Mesothelioma/enzymology , Mice , Mice, Inbred BALB C , Myeloid Cells/pathology , Myeloid Cells/transplantation , Nitric Oxide/metabolism , Reactive Oxygen Species/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/pathology
8.
J Biomed Biotechnol ; 2010: 798467, 2010.
Article in English | MEDLINE | ID: mdl-20508851

ABSTRACT

Clinical immunotherapy trials like dendritic cell-based vaccinations are hampered by the tumor's offensive repertoire that suppresses the incoming effector cells. Regulatory T cells are instrumental in suppressing the function of cytotoxic T cells. We studied the effect of low-dose cyclophosphamide on the suppressive function of regulatory T cells and investigated if the success rate of dendritic cell immunotherapy could be improved. For this, mesothelioma tumor-bearing mice were treated with dendritic cell-based immunotherapy alone or in combination with low-dose of cyclophosphamide. Proportions of regulatory T cells and the cytotoxic T cell functions at different stages of disease were analyzed. We found that low-dose cyclophosphamide induced beneficial immunomodulatory effects by preventing the induction of Tregs, and as a consequence, cytotoxic T cell function was no longer affected. Addition of cyclophosphamide improved immunotherapy leading to an increased median and overall survival. Future studies are needed to address the usefulness of this combination treatment for mesothelioma patients.

10.
Technol Cancer Res Treat ; 8(5): 323-32, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19754208

ABSTRACT

Diagnosis of malignant pleural mesothelioma (MM) is limited. Novel proteomic techno_logies can be utilized to discover changes in expression of pleural proteins that might have diagnostic value. The objective of this study was to detect protein profiles that could be used to identify malignant pleural mesothelioma with surface enhanced laser desorption/ionization time-of-flight (SELDI-TOF) mass spectrometry (MS). Pleural effusions were collected from patients with confirmed mesothelioma (n = 41) and from patients with effusions due to other causes ([n = 48] cancerous and non-cancerous). Samples were fractionated using anion exchange chromatography and bound to different types of ProteinChip array surfaces. All samples were also subjected to other commercially available immunoassays (human epididymes protein 4 [HE4], osteopontin [OPN], soluble mesothelin-related proteins [SMRP], and the cytokeratin 19 fragment [CYFRA 21-1]). Peak intensity data obtained by SELDI-TOF were subjected to classification algorithms in order to identify potential classifier peaks. A protein peak at m/z 6614 was characterized as apolipoprotein (Apo) CI. In this setting, the sensitivity and specificity of this potential biomarker was 76 % and 69 %, respectively. The area under the receiver operating characteristic curve (AUC) for Apo CI was 0.755, thereby outperforming OPN, HE4, and CYFRA 21-1. SMRP performed best with an AUC of 0.860 with a sensitivity of 83% and specificity of 74%. Our study validates the use of SMRP as a diagnostic marker for pleural mesothelioma and furthermore suggests that Apo CI levels could be used in the future to discriminate pleural mesothelioma from other causes of exudates.


Subject(s)
Apolipoprotein C-I/analysis , Biomarkers, Tumor/analysis , Mesothelioma/diagnosis , Neoplasm Proteins/analysis , Pleural Effusion, Malignant/diagnosis , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Adult , Aged , Aged, 80 and over , Area Under Curve , Female , Humans , Immunoassay , Male , Mesothelioma/chemistry , Middle Aged , Neoplasm Staging , Pleural Effusion, Malignant/chemistry , Prognosis , Protein Array Analysis , Proteome/analysis , Sensitivity and Specificity
11.
Methods Mol Biol ; 484: 97-109, 2008.
Article in English | MEDLINE | ID: mdl-18592175

ABSTRACT

Exosomes are small natural membrane vesicles released by a wide variety of cell types into the extracellular compartment by exocytosis. The biological functions of exosomes are only slowly unveiled, but it is clear that they serve to remove unnecessary cellular proteins (e.g., during reticulocyte maturation) and act as intercellular messengers because they fuse easily with the membranes of neighboring cells, delivering membrane and cytoplasmic proteins from one cell to another. Recent findings suggests that cell-derived vesicles (exosomes are also named membranous vesicles or microvesicles) could also induce immune tolerance, suppression of natural killer cell function, T cell apoptosis, or metastasis. For example, by secreting exosomes, tumors may be able to accomplish the loss of those antigens that may be immunogenic and capable of signaling to immune cells as well as inducing dysfunction or death of immune effector cells. On the other hand, dendritic cell-derived exosomes have the potential to be an attractive powerful immunotherapeutic tool combining the antitumor activity of dendritic cells with the advantages of a cell-free vehicle. Although the full understanding of the significance of exosomes requires additional studies, these membrane vesicles could become a new important component in orchestrating responses between cells.


Subject(s)
Exosomes/chemistry , Proteins/analysis , Animals , Cell Line , Electrophoresis, Polyacrylamide Gel/methods , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization/methods
12.
Oncoimmunology ; 7(12): e1474318, 2018.
Article in English | MEDLINE | ID: mdl-30524884

ABSTRACT

Rationale: Regulatory T cells (Treg) play a pivotal role in the immunosuppressive tumor micro-environment in cancer, including mesothelioma. Recently, the combination of autologous tumor lysate-pulsed dendritic cells (DC) and metronomic cyclophosphamide (mCTX) was reported as a feasible and well-tolerated treatment in malignant pleural mesothelioma patients and further as a method to reduce circulating Tregs. Objectives: The aim of this study was to establish the immunological effects of mCTX alone and in combination with DC-based immunotherapy on circulating Treg and other T cell subsets in mesothelioma patients. Methods: Ten patients received mCTX and DC-based immunotherapy after chemotherapy (n = 5) or chemotherapy and debulking surgery (n = 5). Peripheral blood mononuclear cells before, during and after treatment were analyzed for various Treg and other lymphocyte subsets by flow cytometry. Results: After one week treatment with mCTX, both activated FoxP3hi and naïve CD45RA+ Tregs were effectively decreased in all patients. In addition, a shift from naïve and central memory towards effector memory and effector T cells was observed. Survival analysis showed that overall Treg levels before treatment were not correlated with survival, however, nTreg levels before treatment were positively correlated with survival. After completion of mCTX and DC-based immunotherapy treatment, all cell subsets returned to baseline levels, except for the proportions of proliferating EM CD8 T cells, which increased. Conclusions: mCTX treatment effectively reduced the proportions of circulating Tregs, both aTregs and nTregs, thereby favoring EM T cell subsets in mesothelioma patients. Interestingly, baseline levels of nTregs were positively correlated to overall survival upon complete treatment.

13.
Clin Cancer Res ; 24(4): 766-776, 2018 02 15.
Article in English | MEDLINE | ID: mdl-29233904

ABSTRACT

Purpose: Mesothelioma has been regarded as a nonimmunogenic tumor, which is also shown by the low response rates to treatments targeting the PD-1/PD-L1 axis. Previously, we demonstrated that autologous tumor lysate-pulsed dendritic cell (DC) immunotherapy increased T-cell response toward malignant mesothelioma. However, the use of autologous tumor material hampers implementation in large clinical trials, which might be overcome by using allogeneic tumor cell lines as tumor antigen source. The purpose of this study was to investigate whether allogeneic lysate-pulsed DC immunotherapy is effective in mice and safe in humans.Experimental Design: First, in two murine mesothelioma models, mice were treated with autologous DCs pulsed with either autologous or allogeneic tumor lysate or injected with PBS (negative control). Survival and tumor-directed T-cell responses of these mice were monitored. Results were taken forward in a first-in-human clinical trial, in which 9 patients were treated with 10, 25, or 50 million DCs per vaccination. DC vaccination consisted of autologous monocyte-derived DCs pulsed with tumor lysate from five mesothelioma cell lines.Results: In mice, allogeneic lysate-pulsed DC immunotherapy induced tumor-specific T cells and led to an increased survival, to a similar extent as DC immunotherapy with autologous tumor lysate. In the first-in-human clinical trial, no dose-limiting toxicities were established and radiographic responses were observed. Median PFS was 8.8 months [95% confidence interval (CI), 4.1-20.3] and median OS not reached (median follow-up = 22.8 months).Conclusions: DC immunotherapy with allogeneic tumor lysate is effective in mice and safe and feasible in humans. Clin Cancer Res; 24(4); 766-76. ©2017 AACR.


Subject(s)
Allogeneic Cells/immunology , Cancer Vaccines/immunology , Dendritic Cells/immunology , Immunotherapy/methods , Lung Neoplasms/therapy , Mesothelioma/therapy , Aged , Animals , Autografts , Cancer Vaccines/administration & dosage , Cell Extracts/immunology , Cell Line, Tumor , Cells, Cultured , Female , Humans , Lung Neoplasms/immunology , Male , Mesothelioma/immunology , Mesothelioma, Malignant , Mice, Inbred BALB C , Mice, Inbred CBA , Middle Aged , Survival Analysis
14.
Lung Cancer ; 107: 36-40, 2017 05.
Article in English | MEDLINE | ID: mdl-27168021

ABSTRACT

OBJECTIVES: Clinical studies have proven the potential of immunotherapy in malignancies. To increase efficacy, a prerequisite is that treatment is tailored, so precision immune-oncology is the logical next step. In order to tailor treatment, characterization of the patient's tumor environment is key. Pleural effusion (PE) often accompanies malignant pleural mesothelioma (MPM) and is an important part of the MPM environment. Furthermore, the composition of PE is used as surrogate for the tumor. In this study, we provide an insight in the dynamics of the MPM environment through characterization of PE composition over time and show that the immunological characteristics of PE do not necessarily mirror those of the tumor. MATERIALS AND METHODS: From 5 MPM patients, PE and tumor biopsies were acquired at the same time point. From one of these patients multiple PEs were obtained. PEs were acquired performing thoracocenteses and total cell amounts were determined. Immunohistochemistry was performed to quantify immune cell composition (T cells, macrophages) and tumor cells in PE derived cytospins and tumor biopsies. RESULTS: The PE amount and (immune) cellular composition varied considerably over time between multiple (n=10) thoracocenteses. These dynamics could in part be attributed to the treatment regimen consisting of standard chemotherapy and dendritic cell (DC)-based immunotherapy. In addition, the presence of T cells and macrophages in PE did not necessarily mirror the infiltration of these immune cells within tumor biopsies in 4 out of 5 patients. CONCLUSIONS: In this proof-of-concept study with limited sample size, we demonstrate that the composition of PE is dynamic and influenced by treatment. Furthermore, the immune cell composition of PE does not automatically reflect the properties of tumor tissue. This has major consequences when applying precision immunotherapy based on PE findings in patients. Furthermore, it implies a regulated trafficking of immune regulating cells within the tumor environment.


Subject(s)
Immunotherapy/methods , Lung Neoplasms/pathology , Mesothelioma/pathology , Pleural Effusion, Malignant/immunology , Pleural Neoplasms/immunology , Humans , Lung Neoplasms/complications , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Macrophages/metabolism , Mesothelioma/complications , Mesothelioma/drug therapy , Mesothelioma/immunology , Mesothelioma, Malignant , Pleural Effusion, Malignant/pathology , Pleural Effusion, Malignant/therapy , Pleural Neoplasms/drug therapy , Pleural Neoplasms/pathology , Pleural Neoplasms/therapy , Precision Medicine , T-Lymphocytes/immunology , Thoracentesis/methods , Tumor Microenvironment/immunology
15.
Cancer Immunol Res ; 5(7): 535-546, 2017 07.
Article in English | MEDLINE | ID: mdl-28536100

ABSTRACT

New immunotherapeutic strategies are needed to induce effective antitumor immunity in all cancer patients. Malignant mesothelioma is characterized by a poor prognosis and resistance to conventional therapies. Infiltration of tumor-associated macrophages (TAM) is prominent in mesothelioma and is linked to immune suppression, angiogenesis, and tumor aggressiveness. Therefore, TAM depletion could potentially reactivate antitumor immunity. We show that M-CSFR inhibition using the CSF-1R kinase inhibitor PLX3397 (pexidartinib) effectively reduced numbers of TAMs, circulating nonclassical monocytes, as well as amount of neoangiogenesis and ascites in mesothelioma mouse models, but did not improve survival. When combined with dendritic cell vaccination, survival was synergistically enhanced with a concomitant decrease in TAMs and an increase in CD8+ T-cell numbers and functionality. Total as well as tumor antigen-specific CD8+ T cells in tumor tissue of mice treated with combination therapy showed reduced surface expression of the programmed cell death protein-1 (PD-1), a phenomenon associated with T-cell exhaustion. Finally, mice treated with combination therapy were protected from tumor rechallenge and displayed superior T-cell memory responses. We report that decreasing local TAM-mediated immune suppression without immune activation does not improve survival. However, combination of TAM-mediated immune suppression with dendritic cell immunotherapy generates robust and durable antitumor immunity. These findings provide insights into the interaction between immunotherapy-induced antitumor T cells and TAMs and offer a therapeutic strategy for mesothelioma treatment. Cancer Immunol Res; 5(7); 535-46. ©2017 AACR.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive , Lung Neoplasms/immunology , Mesothelioma/immunology , Receptor, Macrophage Colony-Stimulating Factor/immunology , Aminopyridines/administration & dosage , Animals , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Disease Models, Animal , Humans , Lung Neoplasms/chemically induced , Lung Neoplasms/therapy , Macrophages/drug effects , Macrophages/immunology , Mesothelioma/chemically induced , Mesothelioma/therapy , Mesothelioma, Malignant , Neoplasm Invasiveness/immunology , Programmed Cell Death 1 Receptor/immunology , Protein Kinase Inhibitors/administration & dosage , Pyrroles/administration & dosage
16.
J Clin Oncol ; 34(26): 3204-12, 2016 09 10.
Article in English | MEDLINE | ID: mdl-27432922

ABSTRACT

PURPOSE: Programmed cell death protein-1- checkpoint blockers have recently been approved as second-line treatment for advanced non-small-cell lung cancer (NSCLC). Unfortunately, only a subgroup of patients responds and shows long-term survival to these therapies. Tumor vaccines and cellular immunotherapies could synergize with checkpoint blockade, but which of these treatments is most efficacious is unknown. In this meta-analysis, we assessed the efficacy of tumor vaccination and cellular immunotherapy in NSCLC. METHODS: We searched for randomized controlled trials (RCTs) investigating cellular immunotherapy or vaccines in NSCLC. We used random effects models to analyze overall survival (OS) and progression-free survival (PFS), expressed as hazard ratios (HRs), and differences in time (months). The effect of immunotherapy type, disease stage, tumor histology, and concurrent chemotherapy was assessed using subgroup analysis and meta-regression. All procedures were performed according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines. RESULTS: We identified 18 RCTs that matched our selection criteria; these included a total of 6,756 patients. Immunotherapy extended NSCLC survival and PFS, expressed as HR (OS: HR, 0.81, 95% CI, 0.70 to 0.94, P = .01; PFS: HR, 0.83, 95% CI, 0.72 to 0.95, P = .006) and month difference (OS: difference, 5.43 months, 95% CI, 3.20 to 7.65, P < .005; PFS: difference, 3.24 months, 95% CI, 1.61 to 4.88, P < .005). Cellular therapies outperformed tumor vaccines (OS as HR: P = .005, month difference: P < .001; PFS as HR: P = .001, month difference: P = .004). There was a benefit of immunotherapy in low-stage compared with high-stage NSCLC and with concurrent administration of chemotherapy only in one of four outcome measures evaluated (PFS in months: P = .01 and PFS as HR: P = .031, respectively). There was no significant effect of tumor histology on survival or PFS. CONCLUSION: Tumor vaccines and cellular immunotherapies enhanced OS and PFS in NSCLC. Cellular immunotherapy was found to be more effective than tumor vaccination. These findings have implications for future studies investigating combination immunotherapy in NSCLC.


Subject(s)
Cancer Vaccines/therapeutic use , Carcinoma, Non-Small-Cell Lung/therapy , Immunotherapy, Adoptive , Lung Neoplasms/therapy , Cancer Vaccines/adverse effects , Carcinoma, Non-Small-Cell Lung/immunology , Carcinoma, Non-Small-Cell Lung/mortality , Carcinoma, Non-Small-Cell Lung/pathology , Disease-Free Survival , Humans , Immunotherapy, Adoptive/adverse effects , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/pathology , Neoplasm Staging , Risk Factors , Survival Analysis , Time Factors , Treatment Outcome
17.
J Thorac Oncol ; 11(10): 1755-64, 2016 10.
Article in English | MEDLINE | ID: mdl-27418106

ABSTRACT

INTRODUCTION: Clinical studies have demonstrated beneficial effects of immunotherapy in malignant pleural mesothelioma. The pleural cavity seems an attractive compartment to administer these types of therapies; however, local immunosuppressive mechanisms could hamper their efficacy. Macrophages are abundantly present within the mesothelioma microenvironment. This study investigates the influence of the macrophage phenotype, macrophages' capacity to inhibit local immune responses, and the decisive role of pleural effusion (PE) in this regard. METHODS: We cultured macrophages in the presence of PEs and investigated their phenotype. Macrophages and T cells were cocultured in the presence of PEs and tumor cell line supernatants. The levels of 11 cytokines and the prostanoid prostaglandin E2 were measured in PEs and supernatants. The presence and phenotype of macrophages and T cell subsets was measured in the PE of patients with mesothelioma. RESULTS: PE induced a tumor-promoting M2 phenotype in macrophages, which was confirmed by the suppressive activity of macrophages on T cell proliferation during coculture. Prostanoid prostaglandin E2 was identified as a potential inducer of the suppressive capacity of macrophages in PE. Macrophages isolated from PEs displayed an M2 phenotype and were negatively correlated with T cells in vivo. CONCLUSIONS: The current study demonstrates that macrophages in PE can play a pivotal role in directly hampering the antitumor T cell immune response. This emphasizes the potential of macrophages as a therapeutic target in mesothelioma and indicates that the presence and phenotype of macrophages in PE should be taken into consideration in the application of (intrapleural) immunotherapies.


Subject(s)
Lung Neoplasms/complications , Macrophages/metabolism , Mesothelioma/complications , Pleural Effusion/etiology , T-Lymphocytes/pathology , Aged , Cell Line, Tumor , Female , Humans , Lung Neoplasms/pathology , Male , Mesothelioma/pathology , Mesothelioma, Malignant , Pleural Effusion/pathology , Tumor Microenvironment
18.
Lung Cancer ; 88(3): 332-7, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25843042

ABSTRACT

OBJECTIVES: In patients with malignant pleural mesothelioma (MPM), local tumor outgrowth (LTO) after invasive procedures is a well-known complication. Currently, no biomarker is available to predict the occurrence of LTO. This study aims to investigate whether the tumor macrophage infiltration and phenotype of and/or the infiltration of CD8+ T-cells predicts LTO. MATERIALS AND METHODS: Ten mesothelioma patients who developed LTO were clinically and pathologically matched with 10 non-LTO mesothelioma patients. Immunohistochemistry was performed on diagnostic biopsies to determine the total TAM (CD68), the M2 TAM (CD163) and CD8+ T-cell count (CD8). RESULTS: The mean M2/total TAM ratio differed between the two groups: 0.90±0.09 in the LTO group versus 0.63±0.09 in patients without LTO (p<0.001). In addition, the mean CD8+ T-cell count was significantly different between the two groups: 30 per 0.025 cm2 (range 2-60) in the LTO group and 140 per 0.025 cm2 (range 23-314) in the patients without LTO (p<0.01). CONCLUSION: This study shows that patients who develop LTO after a local intervention have a higher M2/total TAM ratio and lower CD8+ cell count at diagnosis compared to patients who did not develop this outgrowth. We propose that the M2/total TAM ratio and the CD8+ T-cell amount are potential tools to predict which MPM patients are prone to develop LTO.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/pathology , Lung Neoplasms/immunology , Lung Neoplasms/pathology , Macrophages/immunology , Macrophages/pathology , Mesothelioma/immunology , Mesothelioma/pathology , Pleural Neoplasms/immunology , Pleural Neoplasms/pathology , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers , Cell Count , Female , Humans , Immunohistochemistry , Lung Neoplasms/drug therapy , Lung Neoplasms/mortality , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/pathology , Macrophages/metabolism , Male , Mesothelioma/drug therapy , Mesothelioma/mortality , Mesothelioma, Malignant , Middle Aged , Neoplasm Staging , Phenotype , Pleural Neoplasms/drug therapy , Pleural Neoplasms/mortality , Prognosis , Tomography, X-Ray Computed , Treatment Outcome
19.
Curr Opin Pharmacol ; 17: 17-21, 2014 Aug.
Article in English | MEDLINE | ID: mdl-25011112

ABSTRACT

We have to strengthen our 'chess-playing skills' when using immunotherapeutic approaches in cancer treatment: know the cancerous opponent, study its evolution and have an eye for its weaknesses. Besides tumor cells, other pieces on the board are stromal cells, endothelial cells and different immune cells. Some of these immune cells, like helper and cytotoxic T cells, natural killer (T) cells and mature dendritic cells are of help, others like regulatory T cells and myeloid-derived suppressor cells belong to the opponent, while macrophages and neutrophils can belong to both. A personalized approach, by selecting the optimal treatment from the myriad of possibilities based on biomarker findings, is essential to attack that particular cancer at that moment. We have to adapt the strategy to the changing positions created by the opponent. Using all these skills, we might control cancer growth in the future.


Subject(s)
Cancer Vaccines/therapeutic use , Immunologic Factors/therapeutic use , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/drug effects , Neoplasms/therapy , Tumor Escape/drug effects , Animals , Biomarkers, Tumor/metabolism , Drug Design , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Molecular Targeted Therapy , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Signal Transduction/drug effects , Tumor Microenvironment
20.
Transl Lung Cancer Res ; 3(1): 34-45, 2014 Feb.
Article in English | MEDLINE | ID: mdl-25806279

ABSTRACT

A very recent finding is the role of immune activation in cancer. The assumption that stimulating the patient's immune system to attack tumors is a valuable treatment option in malignant diseases has gained more acceptance. However the high immunosuppressive effects caused by the tumor limits this beneficial effect. There is a delicate balance between immunoactivation and immunosuppression in a patient. Especially in non small cell lung cancer (NSCLC), the role of immunosuppressive cells hampering immune activation is high. But also in small cell lung cancer (SCLC) and mesothelioma immunosuppressive activity is high. This is suggested to be related to the type of tumor, advanced stage of the disease, and the tumor load. In this review, we provide an overview of the progress and challenges in the immunotherapeutic approaches in lung cancer. We conclude with the concept that immunotherapy in thoracic malignancies must be tailored made to the balance of the immune system.

SELECTION OF CITATIONS
SEARCH DETAIL