Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 138
Filter
Add more filters

Publication year range
1.
J Gen Virol ; 103(1)2022 01.
Article in English | MEDLINE | ID: mdl-35014605

ABSTRACT

The pandemic caused by SARS-CoV-2 has led to the successful development of effective vaccines however the prospect of variants of SARS-CoV-2 and future coronavirus outbreaks necessitates the investigation of other vaccine strategies capable of broadening vaccine mediated T-cell responses and potentially providing cross-immunity. In this study the SARS-CoV-2 proteome was assessed for clusters of immunogenic epitopes restricted to diverse human leucocyte antigen. These regions were then assessed for their conservation amongst other coronaviruses representative of different alpha and beta coronavirus genera. Sixteen highly conserved peptides containing numerous HLA class I and II restricted epitopes were synthesized from these regions and assessed in vitro for their antigenicity against T-cells from individuals with previous SARS-CoV-2 infection. Monocyte derived dendritic cells were generated from these peripheral blood mononuclear cells (PBMC), loaded with SARS-CoV-2 peptides, and used to induce autologous CD4+ and CD8+ T cell activation. The SARS-CoV-2 peptides demonstrated antigenicity against the T-cells from individuals with previous SARS-CoV-2 infection indicating that this approach holds promise as a method to activate anti-SAR-CoV-2 T-cell responses from conserved regions of the virus which are not included in vaccines utilising the Spike protein.


Subject(s)
Peptides/immunology , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Amino Acid Sequence , COVID-19 Vaccines , Coronavirus/classification , Coronavirus/immunology , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA Antigens/immunology , Humans , Leukocytes, Mononuclear/immunology , Lymphocyte Activation , Peptides/chemical synthesis , Peptides/chemistry , Proteome/immunology , Vaccines, Subunit , Viral Proteins/immunology
2.
Immunology ; 161(3): 245-258, 2020 11.
Article in English | MEDLINE | ID: mdl-32794189

ABSTRACT

Release of granulysin by γδ T cells contributes to tumour cell killing. A cytolytic 9000 MW isoform of granulysin kills tumour cells directly, whereas a 15 000 MW precursor has been hypothesized to cause both the maturation and migration of dendritic cell (DC) populations. Recruiting DC to a tumour is beneficial as these cells initiate adaptive immune responses, which contribute to the eradication of malignancies. In this study, Vδ2+ γδ T cells were activated by stimulation of peripheral blood mononuclear cells with zoledronic acid or Bacillus Calmette-Guérin (BCG), or were isolated and cultured with tumour targets. Although a large proportion of resting Vδ2+ γδ T cells expressed 15 000 MW granulysin, 9000 MW granulysin expression was induced only after stimulation with BCG. Increased levels of activation and granulysin secretion were also observed when Vδ2+ γδ T cells were cultured with the human B-cell lymphoma line Daudi. High concentrations of recombinant 15 000 MW granulysin caused migration and maturation of immature DC, and also initiated fugetaxis in mature DC. Conversely, low concentrations of recombinant 15 000 MW granulysin resulted in migration of mature DC, but not immature DC. Our data therefore support the hypothesis that Vδ2+ γδ T cells can release granulysin, which may modulate recruitment of DC, initiating adaptive immune responses.


Subject(s)
Antigens, Differentiation, T-Lymphocyte/metabolism , Dendritic Cells/immunology , Lymphoma, B-Cell/immunology , T-Lymphocytes/immunology , Cell Differentiation , Cell Movement , Cells, Cultured , Chemotaxis , Coculture Techniques , Cytotoxicity, Immunologic , Humans , Lymphocyte Activation , Mycobacterium bovis/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Zoledronic Acid/immunology
3.
J Transl Med ; 16(1): 227, 2018 08 14.
Article in English | MEDLINE | ID: mdl-30107850

ABSTRACT

BACKGROUND: The use of checkpoint inhibitors (ipilimumab, pembrolizumab, nivolumab) has revolutionised the treatment of metastatic melanoma. However still more than the half the patients do not respond to single-agent immunotherapy. This has led to the development of combining these agents in an attempt to enhance the anti-cancer activity. More than 300 different studies with 15 different drug doses are currently ongoing. Combining different checkpoint inhibitors (CPIs) does indeed lead to an increase in response rate, but this is associated with significant toxicity. IMM-101 is a heat killed Mycobacterium preparation which induces marked immune modulation and little systemic toxicity. It has been reported as having activity in melanoma as single agent and in pancreatic cancer in combination with gemcitabine, the latter in a randomised study. METHODS: Here we report the effect of adding CPIs to 3 patients who had previously been on IMM-101, either as a trial or a named patient programme and a patient who received the IMM-101 together with nivolumab. RESULTS: All 4 patients had rapid and very good responses, three of them maintained over 18 months with no significant additional toxicity. CONCLUSIONS: The rapid and complete clinical responses seen in these patients may suggest that IMM-101 is activating a complementary pathway which is synergistic with CPI treatment.


Subject(s)
Cancer Vaccines/adverse effects , Cancer Vaccines/therapeutic use , Immunotherapy , Melanoma/drug therapy , Aged , Female , Humans , Male , Melanoma/diagnostic imaging , Melanoma/pathology , Middle Aged , Neoplasm Staging , Treatment Outcome
4.
Cancer Immunol Immunother ; 66(9): 1205-1215, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28501938

ABSTRACT

Vδ2+ T cells are a subpopulation of γδ T cells in humans that are cytotoxic towards cells which accumulate isopentenyl pyrophosphate. The nitrogen-containing bisphosphonate, zoledronic acid (ZA), can induce tumour cell lines to accumulate isopentenyl pyrophosphate, thus rendering them more susceptible to Vδ2+ T cell cytotoxicity. However, little is known about whether ZA renders other, non-malignant cell types susceptible. In this study we focussed on macrophages (Mϕs), as these cells have been shown to take up ZA. We differentiated peripheral blood monocytes from healthy donors into Mϕs and then treated them with IFN-γ or IL-4 to generate M1 and M2 Mϕs, respectively. We characterised these Mϕs based on their phenotype and cytokine production and then tested whether ZA rendered them susceptible to Vδ2+ T cell cytotoxicity. Consistent with the literature, IFN-γ-treated Mϕs expressed higher levels of the M1 markers CD64 and IL-12p70, whereas IL-4-treated Mϕs expressed higher levels of the M2 markers CD206 and chemokine (C-C motif) ligand 18. When treated with ZA, both M1 and M2 Mϕs became susceptible to Vδ2+ T cell cytotoxicity. Vδ2+ T cells expressed perforin and degranulated in response to ZA-treated Mϕs as shown by mobilisation of CD107a and CD107b to the cell surface. Furthermore, cytotoxicity towards ZA-treated Mϕs was sensitive-at least in part-to the perforin inhibitor concanamycin A. These findings suggest that ZA can render M1 and M2 Mϕs susceptible to Vδ2+ T cell cytotoxicity in a perforin-dependent manner, which has important implications regarding the use of ZA in cancer immunotherapy.


Subject(s)
Bone Density Conservation Agents/therapeutic use , Diphosphonates/therapeutic use , Imidazoles/therapeutic use , Macrophages/metabolism , Animals , Bone Density Conservation Agents/administration & dosage , Bone Density Conservation Agents/pharmacology , Cell Line, Tumor , Cytotoxicity, Immunologic , Diphosphonates/administration & dosage , Diphosphonates/pharmacology , Humans , Imidazoles/administration & dosage , Imidazoles/pharmacology , Mice , Zoledronic Acid
5.
Ann Surg Oncol ; 24(13): 3991-4000, 2017 Dec.
Article in English | MEDLINE | ID: mdl-29019177

ABSTRACT

BACKGROUND: This phase III study was undertaken to evaluate the efficacy of an allogeneic whole-cell vaccine (Canvaxin™) plus bacillus Calmette-Guerin (BCG) after complete resection of stage IV melanoma. METHODS: After complete resection of ≤5 distant metastases, patients were randomly assigned to BCG+Canvaxin (BCG/Cv) or BCG+placebo (BCG/Pl). The primary endpoint was overall survival (OS); secondary endpoints were disease-free survival (DFS), and immune response measured by skin test (ClinicalTrials.gov identifier: NCT00052156). RESULTS: Beginning in May 1998, 496 patients were randomized. In April 2005, the Data Safety Monitoring Board recommended stopping enrollment due to a low probability of efficacy. At that time, median OS and 5-year OS rate were 38.6 months and 44.9%, respectively, for BCG/Pl versus 31.4 months and 39.6% in the BCG/Cv group (hazard ratio (HR), 1.18; p = 0.250). Follow-up was extended at several trial sites through March 2010. Median OS and 5-year and 10-year survival was 39.1 months, 43.3 and 33.3%, respectively, for BCG/Pl versus 34.9 months, 42.5 and 36.4%, in the BCG/Cv group (HR 1.053; p = 0.696). Median DFS, 5- and 10-year DFS were 7.6 months, 23.8 and 21.7%, respectively, for BCG/Pl versus 8.5 months, 30.0%, and 30.0%, respectively, for the BCG/Cv group (HR 0.882; p = 0.260). Positive DTH skin testing correlated with increased survival. DISCUSSION: In this, the largest study of postsurgical adjuvant therapy for stage IV melanoma reported to date, BCG/Cv did not improve outcomes over BCG/placebo. Favorable long-term survival among study patients suggests that metastasectomy should be considered for selected patients with stage IV melanoma.


Subject(s)
Cancer Vaccines/therapeutic use , Immunotherapy/mortality , Melanoma/mortality , Skin Neoplasms/mortality , Combined Modality Therapy , Double-Blind Method , Female , Follow-Up Studies , Humans , Male , Melanoma/pathology , Melanoma/surgery , Melanoma/therapy , Middle Aged , Prognosis , Skin Neoplasms/secondary , Skin Neoplasms/surgery , Skin Neoplasms/therapy , Survival Rate
6.
Future Oncol ; 13(18): 1619-1632, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28776411

ABSTRACT

Immune checkpoint blockades have recently emerged as a breakthrough treatment for solid tumors showing high response rates and long durability. In melanoma, the combination of ipilimumab with nivolumab showed high efficacy. However, still half the patients do not respond to this treatment. In order to increase the therapeutic ratio in melanoma and other cancers, different approaches are under evaluation. Three relevant questions are at the moment driving the research community: how to maximize benefit while minimizing toxicity; how to better identify patients who are more likely to benefit from immunotherapy; how to convert nonresponders into responders. In this review we summarize the most recent findings and we outline the most likely future challenges.


Subject(s)
Antineoplastic Agents, Immunological/therapeutic use , Immunomodulation/drug effects , Neoplasms/immunology , Neoplasms/therapy , Animals , Antineoplastic Agents, Immunological/pharmacology , Antineoplastic Combined Chemotherapy Protocols/adverse effects , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor , Clinical Trials as Topic , Combined Modality Therapy , Humans , Immunotherapy/methods , Molecular Targeted Therapy , Neoplasms/metabolism , Neoplasms/mortality , Treatment Outcome
7.
Br J Cancer ; 115(7): 789-96, 2016 09 27.
Article in English | MEDLINE | ID: mdl-27599039

ABSTRACT

BACKGROUND: Immune Modulation and Gemcitabine Evaluation-1, a randomised, open-label, phase II, first-line, proof of concept study (NCT01303172), explored safety and tolerability of IMM-101 (heat-killed Mycobacterium obuense; NCTC 13365) with gemcitabine (GEM) in advanced pancreatic ductal adenocarcinoma. METHODS: Patients were randomised (2 : 1) to IMM-101 (10 mg ml(-l) intradermally)+GEM (1000 mg m(-2) intravenously; n=75), or GEM alone (n=35). Safety was assessed on frequency and incidence of adverse events (AEs). Overall survival (OS), progression-free survival (PFS) and overall response rate (ORR) were collected. RESULTS: IMM-101 was well tolerated with a similar rate of AE and serious adverse event reporting in both groups after allowance for exposure. Median OS in the intent-to-treat population was 6.7 months for IMM-101+GEM v 5.6 months for GEM; while not significant, the hazard ratio (HR) numerically favoured IMM-101+GEM (HR, 0.68 (95% CI, 0.44-1.04, P=0.074). In a pre-defined metastatic subgroup (84%), OS was significantly improved from 4.4 to 7.0 months in favour of IMM-101+GEM (HR, 0.54, 95% CI 0.33-0.87, P=0.01). CONCLUSIONS: IMM-101 with GEM was as safe and well tolerated as GEM alone, and there was a suggestion of a beneficial effect on survival in patients with metastatic disease. This warrants further evaluation in an adequately powered confirmatory study.


Subject(s)
Antimetabolites, Antineoplastic/therapeutic use , Cancer Vaccines/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Deoxycytidine/analogs & derivatives , Immunotherapy, Active , Pancreatic Neoplasms/drug therapy , Aged , Aged, 80 and over , Antimetabolites, Antineoplastic/administration & dosage , Antimetabolites, Antineoplastic/adverse effects , Biomarkers, Tumor , Cancer Vaccines/administration & dosage , Cancer Vaccines/adverse effects , Carcinoma, Pancreatic Ductal/blood , Carcinoma, Pancreatic Ductal/secondary , Combined Modality Therapy , Deoxycytidine/administration & dosage , Deoxycytidine/adverse effects , Deoxycytidine/therapeutic use , Disease-Free Survival , Europe , Female , Humans , Kaplan-Meier Estimate , Male , Middle Aged , Neoplasm Metastasis , Pancreatic Neoplasms/blood , Pancreatic Neoplasms/pathology , Treatment Outcome , Gemcitabine
8.
Int J Gynecol Cancer ; 26(5): 833-8, 2016 06.
Article in English | MEDLINE | ID: mdl-27120688

ABSTRACT

Mesothelial cells lining the peritoneal cavity are strategically positioned to respond to and counter intraperitoneal infections, cancer cells, and other challenges. We have investigated human peritoneal mesothelial cells (HPMCs) for phagocytic activity, expression of surface Major Histocompatibility Complex (MHC) class II and accessory molecules involved in antigen presentation, and the ability to present recall antigens to T cells. Phagocytosis of dextran, latex beads, and Escherichia coli was observed by flow cytometry, and internalization was visualized using confocal and electron microscopy. Flow cytometry and/or cellular enzyme-linked immunosorbent assay showed constitutive expression of ICAM-1, LFA-3, and B7-1, but not B7-2 or MHC class II. Interferon-gamma induced MHC II and ICAM-1 expression in a dose- and time-dependent manner. Importantly, HPMCs induced autologous CD3 T-lymphocyte proliferation (H incorporation) after pulse with recall antigen. Human peritoneal mesothelial cells equipped with phagocytic and antigen-presenting machinery are anticipated to have an integral role in intraperitoneal immune surveillance.


Subject(s)
Epithelial Cells/immunology , Epithelium/immunology , Antigen Presentation , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Dextrans , Epithelial Cells/cytology , Escherichia coli/immunology , Fluorescein-5-isothiocyanate/analogs & derivatives , Humans , Lymphocyte Activation , Peritoneal Cavity/cytology , Phagocytosis , T-Lymphocytes/immunology
9.
Immunol Rev ; 240(1): 141-59, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21349092

ABSTRACT

As man has moved rapidly from the hunter-gatherer environment to the living conditions of the industrialized countries, the incidences of some cancers have increased alarmingly. Recent increases are usually attributed to dietary changes or to altered exposures to putative carcinogens associated with the modern lifestyle. However, the changes in cancer incidence parallel similar increases in non-neoplastic chronic inflammatory disorders (inflammatory bowel disease, allergies, and autoimmunity), and the epidemiology is often strikingly similar. This parallel is worth exploring, because the increases in chronic inflammatory disorders are at least partly explained by immunoregulatory defects resulting from diminished exposure to microorganisms that co-evolved with mammals and developed a role in driving immunoregulatory circuits (the hygiene hypothesis). Dysregulated chronic inflammation can drive oncogenesis and also provides growth and angiogenic factors that enhance the subsequent proliferation and spread of tumor cells. Thus, a modern failure to downregulate inappropriate inflammation could underlie increases in some cancers in parallel with the increases in chronic inflammatory disorders. This possibility is supported by recent work showing that in some circumstances regulatory T cells protect against cancer, rather than aggravating it, as previously assumed. A greater understanding of these interactions might pave the way to improved microbe-based immunotherapies.


Subject(s)
Bacterial Infections , Neoplasms , Bacterial Infections/immunology , Humans , Immunity , Inflammation , Lymphocytes/immunology , Neoplasms/immunology , Neoplasms/prevention & control
10.
Immunology ; 143(4): 539-49, 2014 Dec.
Article in English | MEDLINE | ID: mdl-24912747

ABSTRACT

Zoledronic acid (ZA) is a potential immunotherapy for cancer because it can induce potent γδ T-cell-mediated anti-tumour responses. Clinical trials are testing the efficacy of intravenous ZA in cancer patients; however, the effects of systemic ZA on the activation and migration of peripheral γδ T cells remain poorly understood. We found that γδ T cells within ZA-treated peripheral blood mononuclear cells were degranulating, as shown by up-regulated expression of CD107a/b. Degranulation was monocyte dependent because CD107a/b expression was markedly reduced in the absence of CD14(+) cells. Consistent with monocyte-induced degranulation, we observed γδ T-cell-dependent induction of monocyte apoptosis, as shown by phosphatidylserine expression on monocytes and decreased percentages of monocytes in culture. Despite the prevailing paradigm that ZA promotes tumour homing in γδ T cells, we observed down-modulation of their tumour homing capacity, as shown by decreased expression of the inflammatory chemokine receptors CCR5 and CXCR3, and reduced migration towards the inflammatory chemokine CCL5. Taken together our data suggest that ZA causes γδ T cells to target monocytes and down-modulate the migratory programme required for inflammatory homing. This study provides novel insight into how γδ T cells interact with monocytes and the possible implications of systemic use of ZA in cancer.


Subject(s)
Diphosphonates/pharmacology , Imidazoles/pharmacology , Inflammation/immunology , Inflammation/metabolism , Monocytes/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/drug effects , T-Lymphocyte Subsets/immunology , Apoptosis/drug effects , Apoptosis/immunology , Cell Degranulation/drug effects , Cell Degranulation/immunology , Cell Line , Cell Movement/drug effects , Cell Movement/immunology , Humans , Immunomodulation/drug effects , Inflammation/drug therapy , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Monocytes/drug effects , Monocytes/metabolism , T-Lymphocyte Subsets/metabolism , Zoledronic Acid
11.
Invest New Drugs ; 32(5): 1048-52, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24442368

ABSTRACT

A role for CD20 antibodies in treating prostate cancer has not yet been established. We report a case of advanced prostate cancer presenting with generalized lymphadenopathy that expressed CCR7 and CD20. CCR7 expression in prostate cancer has been previously reported only once; the expression of CD20 has not been reported before. Rituximab therapy was initiated in this case and resulted in a significant biochemical response. This unique metastatic and biochemical pattern may signify a distinct subtype of prostate cancer that may be amenable to treatment with anti-CD20 antibodies.


Subject(s)
Adenocarcinoma/drug therapy , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antigens, CD20/immunology , Antineoplastic Agents/therapeutic use , Prostatic Neoplasms/drug therapy , Adenocarcinoma/blood , Adenocarcinoma/immunology , Humans , Kallikreins/blood , Lymphatic Diseases/blood , Lymphatic Diseases/drug therapy , Lymphatic Diseases/immunology , Male , Middle Aged , Prostate-Specific Antigen/blood , Prostatic Neoplasms/blood , Prostatic Neoplasms/immunology , Rituximab
12.
World J Surg ; 38(6): 1542-50, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24366277

ABSTRACT

BACKGROUND: The aims of this study were to evaluate the role of surgery in the management of patients with intraabdominal metastases from melanoma and to investigate the neutrophil to lymphocyte ratio (NLR) as prognostic factor in this group of patients. METHODS: Altogether, 44 patients who underwent surgery for Intraabdominal metastases from melanoma with curative, cytoreductive, or palliative intent were analyzed. RESULTS: There were 77 intraabdominal organ resections performed during the 44 operations. R0 resection was achieved in 19 (43 %) cases. Factors associated with R0 resection were an absence of extra-abdominal metastases, low serum lactate dehydrogenase, involvement of fewer than three sites, and the presence of fewer than three metastatic lesions. The 1-, 3-, and 5-year overall survival rates were, respectively, 79, 66, and 44 % in the curative intent group; 36, 18, and 9 % in the cytoreduction group; and 21, 0 and 0 % in the palliation group (curative intent vs. cytoreduction vs. palliation p < 0.001). By Cox's multivariate analysis, the independent prognostic factors were time from excision of primary melanoma to the diagnosis of intraabdominal metastases, NLR, and residual disease after surgery. CONCLUSIONS: Our results confirm the usefulness of major surgical interventions as reported in previous studies. We reviewed recent evidence that immunologic phenomena may explain the unexpectedly good response rate in patients with advanced disease. The simple estimation of the NLR has been advocated as a prognostic marker for several cancers. We show that it is likewise useful in metastatic melanoma. We stress the need for developing additional immunologic markers.


Subject(s)
Biomarkers, Tumor/blood , Gastrointestinal Neoplasms/secondary , Gastrointestinal Neoplasms/surgery , Lymphocyte Count , Melanoma/secondary , Neutrophils/metabolism , Abdominal Neoplasms/mortality , Abdominal Neoplasms/secondary , Abdominal Neoplasms/surgery , Adult , Aged , Aged, 80 and over , Analysis of Variance , Cohort Studies , Databases, Factual , Disease-Free Survival , Female , Gastrointestinal Neoplasms/mortality , Humans , Male , Melanoma/mortality , Melanoma/pathology , Melanoma/surgery , Middle Aged , Prognosis , Risk Assessment , Sensitivity and Specificity , Skin Neoplasms/mortality , Skin Neoplasms/pathology , Skin Neoplasms/surgery , Survival Rate , Treatment Outcome , Young Adult
13.
Immunology ; 139(3): 377-85, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23374145

ABSTRACT

The IMiDs(®) immunomodulatory compounds lenalidomide and pomalidomide are agents with anti-inflammatory, immunomodulatory and anti-cancer activity. An excellent success rate has been shown for multiple myeloma in phase I/II clinical trials leading to Food and Drug Administration approval of lenalidomide. One mechanism by which these drugs could enhance anti-tumour immunity may be through enhanced dendritic cell (DC) function. Thalidomide, a compound structurally related to lenalidomide and pomalidomide, is known to enhance DC function, and we have investigated whether its analogues, pomalidomide and lenalidomide, also have functional effects on DCs. We used mouse bone marrow-derived DCs treated with 5 or 10 µm pomalidomide, or lenalidomide from day 1 of culture. Treatment with IMiD(®) immunomodulatory compounds increased expression of Class I (H2-Kb), CD86, and pomalidomide also increased Class II (I-Ab) expression in bone marrow-derived DCs, as measured by flow cytometry. Fluorescent bead uptake was increased by up to 45% when DCs were treated with 5 or 10 µm pomalidomide or lenalidomide compared with non-treated DCs. Antigen presentation assays using DCs primed with ovalbumin, and syngeneic T cells from transgenic OTI and OTII mice (containing MHC restricted, ovalbumin-specific, T cells) showed that both pomalidomide and lenalidomide effectively increased CD8(+) T-cell cross-priming (by up to 47%) and that pomalidomide alone was effective in increasing CD4(+) T-cell priming (by 30%). Our observations suggest that pomalidomide and lenalidomide enhance tumour antigen uptake by DCs with an increased efficacy of antigen presentation, indicating a possible use of these drugs in DC vaccine therapies.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/drug effects , Thalidomide/analogs & derivatives , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Antigens, Neoplasm/immunology , Cells, Cultured , Dendritic Cells/immunology , Female , Immunologic Factors/immunology , Immunologic Factors/pharmacology , Lenalidomide , Lymphocyte Activation/drug effects , Mice , Mice, Inbred C57BL , Ovalbumin/immunology , Thalidomide/immunology , Thalidomide/pharmacology
15.
Cancer Immunol Immunother ; 62(10): 1553-61, 2013 Oct.
Article in English | MEDLINE | ID: mdl-23824498

ABSTRACT

INTRODUCTION: Dendritic cells (DCs) possess the capacity to elicit immune responses against harmful antigens and have been used in DC-vaccines to stimulate the immune system to engage cancer cells. However, a lack of an appreciation of the quality of the DC that is used and/or the monocyte from which it is derived has limited their successful incorporation into treatment strategies. METHODS: In the current study, we explored the relationship between cytokine receptor expression on the monocytes and its subsequent development into DCs. The significance of p21 expression in DCs during differentiation was also studied, as was the effect that manipulating this with chemotherapy may have on DC quality. RESULTS: DCs separated into two groups based on their ability to respond to a maturation stimulus. This quality correlated with a particular receptor profile of granulocyte-macrophage colony-stimulating factor and interleukin 4 expressed on the monocytes from which they were derived. DC quality was also associated with p21 expression, and artificially increasing their levels in DCs by using some chemotherapy improved function. CONCLUSIONS: Overall, these studies have highlighted a role for common chemotherapy in activating p21 in DCs, which is a prerequisite for good DC function.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p21/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Cell Differentiation/immunology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology , Humans , Immunotherapy , Interleukin-4/pharmacology , Monocytes/cytology , Monocytes/drug effects , Monocytes/immunology , Phenotype
16.
Cancer Immunol Immunother ; 62(7): 1223-33, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23624802

ABSTRACT

We analysed mRNA levels of interferon response genes (ISG15, STAT1, CXCL10) of inhibitors of the JAK/STAT pathway (STAT3, SOCS1, SOCS3) and of cytokines (TNFα, IL10, TGFß1) in peripheral blood of 91 stage III melanoma patients enrolled in EORTC 18991 trial to find biomarkers indicative for disease stage and predictive for efficacy of pegylated interferon alpha-2b (PEG-IFNα-2b) therapy. mRNA levels were analysed at baseline and after 6 months. Univariate and multivariate analyses were performed to estimate the prognostic and predictive role of mRNA levels for distant metastasis-free survival (DMFS) and relapse-free survival (RFS). Compared to healthy controls, melanoma patients showed significantly higher TGFß1 mRNA levels. In a multivariate model, increasing SOCS1 and SOCS3 mRNA levels were associated with worse RFS (P = 0.02 and P = 0.04, respectively) and DMFS (P = 0.05 and P = 0.05, respectively) due to negative correlation between, respectively, SOCS1/SOCS3 mRNA levels and ulceration or Breslow thickness. No impact of PEG-IFNα-2b on mRNA levels was observed except for ISG15 mRNA levels, which decreased in the treatment arm (P = 0.001). It seems that patients with a decrease >60 % of ISG15 mRNA levels during 6 months PEG-IFNα-2b had inferior outcome.


Subject(s)
Antineoplastic Agents/therapeutic use , Cytokines/genetics , Interferon-alpha/therapeutic use , Melanoma/drug therapy , Polyethylene Glycols/therapeutic use , RNA, Messenger/blood , Skin Neoplasms/drug therapy , Adult , Biomarkers, Tumor/genetics , Chemotherapy, Adjuvant , Cytokines/blood , Disease-Free Survival , Female , Gene Expression , Humans , Interferon alpha-2 , Male , Melanoma/blood , Melanoma/genetics , Melanoma/mortality , Middle Aged , Recombinant Proteins/therapeutic use , Skin Neoplasms/mortality , Treatment Outcome
17.
Immunol Cell Biol ; 91(7): 461-8, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23797069

ABSTRACT

γδ T cells contribute to immunosurveillance of pathogenic infections and malignant transformations; however, mechanisms of activation have yet to be fully defined. In this study we demonstrate a novel mechanism by which human Vδ2(+) γδ T cells are activated by the model pathogen Bacillus Calmette Guérin (BCG). We show in vitro that Vδ2 cell cytokine production and cytotoxic activity in response to BCG are dependent on both dendritic cells (DCs) and memory CD4(+) αß T cells (CD4 T cells). We found that Vδ2 cells are indirectly activated by BCG in an interleukin (IL)-12p70-dependent manner, and that DC production of the IL-12p70 responsible for Vδ2 cell activation requires Toll-like receptor 2/4 ligands from BCG and interferon (IFN)-γ from memory CD4 T cells. Our data suggest that Vδ2 cell responses to BCG are dependent on the activation of IFN-γ-producing memory CD4 T cells, and provide novel insight into the complex interplay between cells of the innate and adaptive immune response.


Subject(s)
BCG Vaccine/immunology , CD4-Positive T-Lymphocytes/immunology , T-Lymphocytes, Cytotoxic/immunology , Cell Communication , Cells, Cultured , Cytotoxicity, Immunologic , Humans , Immunologic Memory , Interferon-gamma/metabolism , Interleukin-12/metabolism , Lymphocyte Activation , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism
18.
BMC Cancer ; 13: 41, 2013 Jan 31.
Article in English | MEDLINE | ID: mdl-23367880

ABSTRACT

BACKGROUND: Aberrant expression and activation of the IGF-IR have been reported in a variety of human cancers and have been associated with resistance to HER targeted therapy. In this study, we investigated the effect of simultaneous targeting of IGF-IR and HER (erbB) family, with NVP-AEW541 and afatinib, on proliferation of pancreatic cancer cells. METHODS: The sensitivity of a panel of human pancreatic cancer cell lines to treatment with NVP-AEW541 used alone or in combination with afatinib, anti-EGFR antibody ICR62, and cytotoxic agents was determined using the Sulforhodamine B colorimetric assay. Growth factor receptor expression, cell-cycle distribution and cell signalling were determined using flow cytometry and western blot analysis. RESULTS: All pancreatic cancer cell lines were found to be IGF-IR positive and NVP-AEW541 treatment inhibited the growth of the pancreatic cancer cell lines with IC50 values ranging from 342 nM (FA6) to 2.73 µM (PT45). Interestingly, of the various combinations examined, treatment with a combination of NVP-AEW541 and afatinib was superior in inducing synergistic growth inhibition of the majority of pancreatic cancer cells. CONCLUSION: Our results indicate that co-targeting of the erbB (HER) family and IGF-IR, with a combination of afatinib and NVP-AEW541, is superior to treatment with a single agent and encourages further investigation in vivo on their therapeutic potential in IGF-IR and HER positive pancreatic cancers.


Subject(s)
Antineoplastic Agents/pharmacology , Pancreatic Neoplasms/drug therapy , Pyrimidines/pharmacology , Pyrroles/pharmacology , Quinazolines/pharmacology , Receptor, ErbB-3/antagonists & inhibitors , Receptor, IGF Type 1/antagonists & inhibitors , Afatinib , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Therapy, Combination/methods , Humans , Pancreatic Neoplasms/metabolism , Receptor, IGF Type 1/metabolism
19.
Int J Cancer ; 130(4): 979-90, 2012 Feb 15.
Article in English | MEDLINE | ID: mdl-21455985

ABSTRACT

Dendritic cells (DCs) represent a subset of professional antigen presenting cell (APC) whose role is to elicit immune responses against harmful antigens. They have been used in DC vaccines to stimulate the immune system to kill cancer cells. However, successes in clinical trials have been limited, which may be attributed to a lack of appreciation of the quality of DCs used. In the present study, whole human genome microarrays were used to examine alterations in gene expression of monocyte-derived DCs after stimulation with supernatants derived from tumours. Our primary aim was to investigate the possibility of a gene signature for DCs that could be used to forecast responsiveness to tumour stimuli. Results showed that DCs are divided into two groups based on their ability to increase costimulatory markers and to trigger T-cell responses. The gene profiles of the immature DCs from these two groups were distinct, with particular divergence in genes from the interleukin (IL) 8 and thrombospondin-1 hubs. A subpanel of genes was identified, whose signature of expression was capable of predicting DC-stimulatory capacity. Overall, these studies have highlighted a gene-based screen that predicts DC function, which could be used to guide DC-vaccine trials.


Subject(s)
Dendritic Cells/immunology , Gene Expression Profiling , Cell Line, Tumor , Dendritic Cells/metabolism , Humans , Neoplasms/immunology , Neoplasms/therapy , Oligonucleotide Array Sequence Analysis , Vaccines/immunology
20.
Apoptosis ; 17(2): 164-73, 2012 Feb.
Article in English | MEDLINE | ID: mdl-22109882

ABSTRACT

1α,25-Dihydroxyvitamin D3, (1,25-D3) the biologically active form of vitamin-D, is well established as a cancer cell growth inhibitor in addition to maintaining bone mineralization. In breast cancer cells, inhibitory effects on angiogenesis, and metastasis have been observed together with enhancement of apoptosis and induction of cell cycle arrest. There is a correlation between vitamin-D receptor expression on breast cancer cells and patient survival. However vitamin-D resistance and hypercalcaemia are key limiting factors in clinical use. The IMiD(®) immunomodulatory drug lenalidomide, (Revlimid(®), CC-5013) used in myeloma, can also modulate apoptotic and growth signalling. We studied whether lenalidomide treated breast cancer cells would acquire sensitivity to 1,25-D3 with resulting growth inhibition. The cell lines MCF-12A, MCF-7 and MDA-MB-231, representing non-tumorogenic, tumorogenic, and vitamin-D resistant lines respectively were treated with lenalidomide and/or 1,25-D3(at 100 nM). Whereas lenalidomide alone had no effect on cell growth, a 50% inhibition of cell growth by 1,25-D3 was achieved with additional 1 µM lenalidomide in resistant cells. This effect was through apoptosis measured by PARP cleavage and annexin-V expression. An apoptosis protein array showed that the 1,25-D3 and lenalidomide combination increased pro-apoptotic proteins (phosphorylated p53) and decreased BCL-2 expression. BCL-2 inhibition is proposed as a mechanism of action for the combined drugs in the MDA-MB-231 cell line. In vitamin D resistant cell lines MCF-7VDR and HBL-100 where the combination does not affect BCL-2-no inhibitory effect is observed. These results demonstrate the potential for the combinatorial use of lenalidomide and 1,25-D3 for vitamin D refractory tumours.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/administration & dosage , Breast Neoplasms/metabolism , Calcitriol/administration & dosage , Thalidomide/analogs & derivatives , Apoptosis/drug effects , Breast Neoplasms/therapy , Cell Line, Tumor , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lenalidomide , Phosphorylation/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Receptors, Calcitriol/genetics , Receptors, Calcitriol/metabolism , Thalidomide/administration & dosage , Tumor Suppressor Protein p53/genetics , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL