Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 7(1): e1376154, 2017.
Article in English | MEDLINE | ID: mdl-29296535

ABSTRACT

Vaccine-mediated cancer treatment is unlikely to induce long-term survival unless suppressive mechanisms are overcome. Given the success of antibody-mediated immune checkpoint blockade in relieving regulation of endogenous anti-tumor T cell responses in tumor-burdened hosts, we investigated whether checkpoint blockade could improve the efficacy of responses induced with a whole tumor-cell vaccine. We show that administration of a single dose of blocking antibody was sufficient to significantly enhance antitumor activity of the vaccine, inducing complete radiological regression of established intracranial tumors. The antibody or vaccine alone were ineffective in this setting. The antibody had to be administered before, or close to, vaccine administration, suggesting CTLA-4 blockade had an impact on early priming events. The combined treatment resulted in enhanced trapping of leukocytes in the lymphoid tissues, including T cells that had undergone significant proliferation. There were no obvious changes in the stimulatory function of antigen-presenting cells or the number and function of regulatory T cells, suggesting T cells were the targets of the checkpoint blockade. While tumors regressing under combined treatment were highly infiltrated with a variety of leukocytes, tumor eradication was dependent on CD4+ T cells. Analysis of the TCR repertoire showed that the addition of anti-CTLA-4 at priming reshaped the repertoire of tumor infiltrating T cells. In particular, the oligoclonal populations became greater in magnitude and more diverse in specificity. Using anti-CTLA-4 in a restricted way to promote the priming phase of an anti-cancer vaccine may offer a useful way of harnessing clinical benefit from this powerful agent.

2.
Magn Reson Chem ; 55(5): 498-507, 2017 May.
Article in English | MEDLINE | ID: mdl-27487091

ABSTRACT

Complex materials are ubiquitous in science, engineering and nature. One important parameter for characterising their morphology is the degree of anisotropy. Magnetic resonance imaging offers non-invasive methods for quantitative measurements of the materials anisotropy, most commonly via diffusion tensor imaging and the subsequent extraction of the spatially resolved fractional anisotropy (FA) value. Here, we propose an alternative way of determining the FA as a sample average for cases where spatially resolved methods are not needed or not applicable. It is based on a particular diffusion-diffusion correlation spectroscopy protocol, allowing for the extraction of the mean (i.e. sample averaged) FA value. We demonstrate that mean FA values obtained from three anisotropic biological tissues are consistent with those extracted using diffusion tensor imaging. Moreover, we show that differences of mean FA values in healthy and tumour-bearing mouse brains allow to distinguish these tissue types. We anticipate that the proposed method will be beneficial in the wider context of medical and material science. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Magnetic Resonance Spectroscopy/methods , Animals , Anisotropy , Brain/diagnostic imaging , Brain Neoplasms/diagnostic imaging , Cell Line, Tumor , Computer Simulation , Daucus carota , Diffusion , Diffusion Tensor Imaging , Humans , Mice
3.
J Immunother ; 38(8): 321-9, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26325376

ABSTRACT

Circulating antigens released from tumor cells can drain into the spleen and be acquired by resident antigen-presenting cells (APCs). Here, we examined the ability of splenic dendritic cells to cross-present tumor antigens to CD8+ T cells and investigated the effects that this has on T-cell therapy in a murine model of lymphoma. In the presence of established lymphoma, langerin (CD207)-expressing CD8α+ dendritic cells acquired, processed, and cross-presented tumor antigens to naive CD8+ T cells. Although this resulted in initial T-cell proliferation, the T-cell population failed to expand measurably over the following days, and tumor-free survival was actually improved when langerin-expressing cells were depleted. In contrast, following adoptive T-cell therapy with in vitro-activated CD8+ T cells, marked antitumor activity was observed and associated with accumulation of activated antigen-specific CD8+ T cells in the spleen and blood, whereas tumor protection and T-cell accumulation were significantly reduced in animals depleted of langerin-expressing cells. Therefore, although resident APCs that acquire tumor antigens may induce tolerance in naive cells in the absence of further stimuli, they can play an important role in promoting antitumor immunity during the course of T-cell therapy. It is possible that further therapeutic benefit will result from improving the activation status of these APCs.


Subject(s)
Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive , Animals , Antigens, Surface/immunology , Cell Line, Tumor , Lectins, C-Type/immunology , Lymphoma/therapy , Mannose-Binding Lectins/immunology , Mice, Inbred C57BL , Mice, Transgenic , Spleen/cytology , Spleen/immunology
4.
Oncoimmunology ; 4(4): e995568, 2015 Apr.
Article in English | MEDLINE | ID: mdl-26137409

ABSTRACT

Many acute leukemia patients treated with chemotherapy are at high risk of relapse without allogeneic stem cell transplantation, an immunotherapy that is limited by significant toxicity and donor availability. We propose that post-remission vaccination with a simple autologous whole cell vaccine adjuvanted with α-galactosylceramide may be effective to prevent relapse of acute leukemia.

5.
J Neurooncol ; 121(2): 319-29, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25366363

ABSTRACT

There is no standard treatment for recurrent glioblastoma multiforme (GBM). Retreatment with temozolomide (TMZ) is one treatment option. We reasoned this could be more effective if combined with a vaccine that preferentially targeted TMZ-resistant cells. To test the feasibility and safety of such an approach, a phase 1 trial was conducted in which patients with GBM tumors relapsing after standard chemoradiotherapy were retreated with TMZ in combination with a vaccine consisting of monocyte-derived dendritic cells (DC) pulsed with autologous tumor cells that had previously been exposed to TMZ in vivo in the course of primary treatment. Of 14 participants, nine patients completed the initial phase of priming vaccinations and two cycles of TMZ, one proved to have radionecrosis, one rapidly progressed, and in three the yield of DC vaccine was insufficient to proceed with treatment. Other than expected toxicities related to TMZ, there were no adverse events attributable to the combined treatment. Two patients had objective radiological responses. Six month progression-free survival was 22 %, similar to retreatment with TMZ alone. Anti-tumor immune responses were assessed in peripheral blood mononuclear cells using interferon-γ ELISpot, with two patients meeting criteria for a vaccine-induced immune response, one of whom remained disease-free for nearly three years. Another patient with an anti-tumor immune response at baseline that was sustained post-vaccination experienced a 12-month period of progression-free survival. In summary, the combined treatment was safe and well-tolerated but feasibility in the recurrent setting was marginal. Evidence of immune responses in a few patients broadly correlated with better clinical outcome.


Subject(s)
Antineoplastic Agents, Alkylating/therapeutic use , Brain Neoplasms/therapy , Cancer Vaccines/therapeutic use , Dacarbazine/analogs & derivatives , Glioblastoma/therapy , Neoplasm Recurrence, Local/therapy , Adult , Aged , Brain/pathology , Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cancer Vaccines/adverse effects , Combined Modality Therapy/adverse effects , Dacarbazine/adverse effects , Dacarbazine/therapeutic use , Dendritic Cells/immunology , Disease-Free Survival , Feasibility Studies , Female , Glioblastoma/immunology , Glioblastoma/pathology , Humans , Magnetic Resonance Imaging , Male , Middle Aged , Neoplasm Recurrence, Local/immunology , Neoplasm Recurrence, Local/pathology , Retreatment , Temozolomide , Treatment Outcome
6.
Int J Cancer ; 136(11): 2566-78, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25363661

ABSTRACT

Glioblastoma multiforme (GBM) is a highly malignant brain tumor with an extremely short time to relapse following standard treatment. Since recurrent GBM is often resistant to subsequent radiotherapy and chemotherapy, immunotherapy has been proposed as an alternative treatment option. Although it is well established that GBM induces immune suppression, it is currently unclear what impact prior conventional therapy has on the ability of GBM cells to modulate the immune environment. In this study, we investigated the interaction between immune cells and glioma cells that had been exposed to chemotherapy or irradiation in vitro. We demonstrate that treated glioma cells are more immunosuppressive than untreated cells and form tumors at a faster rate in vivo in an animal model. Cultured supernatant from in vitro-treated primary human GBM cells were also shown to increase suppression, which was independent of accessory suppressor cells or T regulatory cell generation, and could act directly on CD4(+) and CD8(+) T cell proliferation. While a number of key immunosuppressive cytokines were overexpressed in the treated cells, including IL-10, IL-6 and GM-CSF, suppression could be alleviated in a number of treated GBM lines by inhibition of prostaglandin E2. These results reveal for the first time that conventional therapies can alter immunosuppressive pathways in GBM tumor cells, a finding with important implications for the combination of immunotherapy with standard treatment.


Subject(s)
Brain Neoplasms/immunology , Brain Neoplasms/pathology , Cytokines/metabolism , Glioblastoma/immunology , Glioblastoma/pathology , Animals , Brain Neoplasms/therapy , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Proliferation , Culture Media, Conditioned/pharmacology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , Glioblastoma/therapy , Humans , Immunosuppression Therapy , Mice , Mice, Inbred C57BL , Neoplasm Transplantation
7.
Blood ; 124(19): 2953-63, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25237205

ABSTRACT

Acute leukemias with adverse prognostic features carry a high relapse rate without allogeneic stem cell transplantation (allo-SCT). Allo-SCT has a high morbidity and is precluded for many patients because of advanced age or comorbidities. Postremission therapies with reduced toxicities are urgently needed. The murine acute leukemia model C1498 was used to study the efficacy of an intravenously administered vaccine consisting of irradiated leukemia cells loaded with the natural killer T (NKT)-cell agonist α-galactosylceramide (α-GalCer). Prophylactically, the vaccine was highly effective at preventing leukemia development through the downstream activities of activated NKT cells, which were dependent on splenic langerin(+)CD8α(+) dendritic cells and which led to stimulation of antileukemia CD4(+) and CD8(+) T cells. However, hosts with established leukemia received no protective benefit from the vaccine, despite inducing NKT-cell activation. Established leukemia was associated with increases in regulatory T cells and myeloid-derived suppressor cells, and the leukemic cells themselves were highly suppressive in vitro. Although this suppressive environment impaired both effector arms of the immune response, CD4(+) T-cell responses were more severely affected. When cytarabine chemotherapy was administered prior to vaccination, all animals in remission posttherapy were protected against rechallenge with viable leukemia cells.


Subject(s)
Cancer Vaccines/pharmacology , Cytarabine/pharmacology , Galactosylceramides/immunology , Killer Cells, Natural/transplantation , Leukemia, Myeloid/drug therapy , Leukemia, Myeloid/prevention & control , Acute Disease , Animals , Antimetabolites, Antineoplastic/pharmacology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Combined Modality Therapy , Dendritic Cells/immunology , Green Fluorescent Proteins/genetics , Killer Cells, Natural/radiation effects , Leukemia, Myeloid/immunology , Mice, Inbred C57BL , Mice, Transgenic , Prognosis , Secondary Prevention/methods , Transplantation, Autologous
8.
PLoS One ; 7(9): e45888, 2012.
Article in English | MEDLINE | ID: mdl-23049885

ABSTRACT

Oral delivery of BCG in a lipid formulation (Liporale™-BCG) targets delivery of viable bacilli to the mesenteric lymph nodes and confers protection against an aerosol Mycobacterium tuberculosis challenge. The magnitude, quality and duration of the effector and memory immune response induced by Liporale™-BCG vaccination is unknown. Therefore, we compared the effector and memory CD4(+) T cell response in the spleen and lungs of mice vaccinated with Liporale™-BCG to the response induced by subcutaneous BCG vaccination. Liporale™-BCG vaccination induced a long-lived CD4(+) T cell response, evident by the detection of effector CD4(+) T cells in the lungs and a significant increase in the number of Ag85B tetramer-specific CD4(+) T cells in the spleen up to 30 weeks post vaccination. Moreover, following polyclonal stimulation, Liporale™-BCG vaccination, but not s.c. BCG vaccination, induced a significant increase in both the percentage of CD4(+) T cells in the lungs capable of producing IFNγ and the number of multifunctional CD4(+) T cells in the lungs at 30 weeks post vaccination. These results demonstrate that orally delivered Liporale™-BCG vaccine induces a long-lived multifunctional immune response, and could therefore represent a practical and effective means of delivering novel BCG-based TB vaccines.


Subject(s)
BCG Vaccine/therapeutic use , CD4-Positive T-Lymphocytes/immunology , Vaccines/therapeutic use , Acyltransferases/chemistry , Administration, Oral , Animals , Antigens, Bacterial/chemistry , Bacterial Proteins/chemistry , Female , Immune System , Interferon-gamma/metabolism , Lipids/chemistry , Lung/immunology , Male , Mice , Mice, Inbred C57BL , Mycobacterium tuberculosis/metabolism , Spleen/cytology
9.
Tuberculosis (Edinb) ; 92(5): 422-33, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22738879

ABSTRACT

Several studies have used adoptive transfer of purified T cell subsets into immunodeficient mice to determine the subset of T cells responsible for mediating protection against Mycobacterium tuberculosis. These studies suggested that CD62L(hi) memory CD4(+) T cells from BCG-vaccinated mice are key for protection against tuberculosis. Importantly, we observed that transfer of naïve CD4(+) T cells into Rag1-/- recipients protected against a mycobacterial challenge as well as transfer of BCG-experienced CD4(+) T cells. We found that transfer of total CD4(+) T cells from naïve mice or enriched CD62L(hi)CD4(+) T cells from BCG-vaccinated mice into Rag1-/- recipients induced severe colitis by 3 weeks post cell transfer, whereas transfer of CD62L(lo)CD4(+) T cells from BCG-vaccinated mice did not. Naïve and CD62L(hi)CD4(+) T cells proliferated extensively upon transfer and developed an activated effector phenotype in the lung, even in the absence of infectious challenge. The induction of colitis and systemic cytokine response induced by the transfer and subsequent activation of CD4(+) T cells from naïve mice or CD62L(hi)CD4(+) T cells from BCG-vaccinated mice, into immunodeficient recipients, may heighten their ability to protect against mycobacterial challenge. This raises doubts about the validity of this model to study CD4(+) T cell-mediated protection against tuberculosis.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Colitis/immunology , Colon/immunology , Mycobacterium bovis/immunology , Mycobacterium tuberculosis/immunology , Tuberculosis/immunology , Adoptive Transfer , Animals , Antigens, Bacterial/immunology , Colitis/pathology , Colon/pathology , Flow Cytometry , Homeodomain Proteins/immunology , Immunologic Memory , Mice , Mice, Inbred C57BL , Models, Immunological , Mycobacterium bovis/pathogenicity , Mycobacterium tuberculosis/pathogenicity , Tuberculosis/pathology , Vaccination
10.
Biomol Concepts ; 3(1): 13-20, 2012 Feb.
Article in English | MEDLINE | ID: mdl-25436521

ABSTRACT

Abstract Effective vaccination against intracellular pathogens, such as tuberculosis (TB), relies on the generation and maintenance of CD4 memory T cells. An incomplete understanding of the memory immune response has hindered the rational design of a new, more effective TB vaccine. This review discusses how the persistence of antigen, the location of memory cells, and their multifunctional ability shape the CD4 memory T cell response against TB.

SELECTION OF CITATIONS
SEARCH DETAIL
...