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










Publication year range
1.
Int J Cancer ; 143(9): 2200-2212, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29752716

ABSTRACT

Solid tumors, including gliomas, still represent a challenge to clinicians and first line treatments often fail, calling for new paradigms in cancer therapy. Novel strategies to overcome tumor resistance are mainly represented by multi-targeted approaches, and cell vector-based therapy is one of the most promising treatment modalities under development. Here, we show that mouse bone marrow-derived mesenchymal stromal cells (MSCs), when primed with low-dose irradiation (irMSCs), undergo changes in their immunogenic and angiogenic capacity and acquire anti-tumoral properties in a mouse model of glioblastoma (GBM). Following grafting in GL261 glioblastoma, irMSCs migrate extensively and selectively within the tumor and infiltrate predominantly the peri-vascular niche, leading to rejection of established tumors and cure in 29% of animals. The therapeutic radiation dose window is narrow, with effects seen between 2 and 15 Gy, peaking at 5 Gy. A single low-dose radiation decreases MSCs inherent immune suppressive properties in vitro as well as shapes their immune regulatory ability in vivo. Intra-tumorally grafted irMSCs stimulate the immune system and decrease immune suppression. Additionally, irMSCs enhance peri-tumoral reactive astrocytosis and display anti-angiogenic properties. Hence, the present study provides strong evidence for a therapeutic potential of low-dose irMSCs in cancer as well as giving new insight into MSC biology and applications.


Subject(s)
Glioblastoma/immunology , Glioblastoma/therapy , Mesenchymal Stem Cell Transplantation , Mesenchymal Stem Cells/immunology , Animals , Apoptosis , Cell Proliferation , Female , Glioblastoma/blood supply , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/radiation effects , Mice , Mice, Inbred C57BL , Tumor Cells, Cultured
2.
J Neurooncol ; 131(2): 245-254, 2017 01.
Article in English | MEDLINE | ID: mdl-27757723

ABSTRACT

Gene profiling has revealed that malignant gliomas can be divided into four distinct molecular subtypes, where tumors with a mesenchymal gene expression are correlated with short survival. The present investigation was undertaken to clarify whether human malignant gliomas contain endogenous mesenchymal stromal cells (MSC), fulfilling consensus criteria defined by The International Society for Cellular Therapy, recruited from the host. We found that MSC-like cells can be isolated from primary human malignant gliomas. Two distinct MSC-like cell populations, differing in their expression of the CD90 surface marker, were discovered after cell sorting. RNA sequencing revealed further genetic differences between these two cell populations and MSC-like cells lacking CD90 produced higher amounts of VEGF and PGE2 compared to cells with the true MSC phenotype, implying that the CD90- MSC-like cells most probably are more active in tumor vascularization and immunosuppression than their CD90+ counterpart. The results highlight the CD90- subpopulation as an important tumor component, however, its functional effects in glioma remains to be resolved. Using the protocols presented here, it will be possible to isolate, characterize and analyze brain tumor-derived MSC-like cells in more detail and to further test their functions in vitro and in in vivo xenograft models of glioma.


Subject(s)
Brain Neoplasms/pathology , Glioma/pathology , Mesenchymal Stem Cells/pathology , Adult , Aged , Brain Neoplasms/genetics , Dinoprostone , Female , Gene Expression Profiling , Glioma/genetics , Humans , Low Density Lipoprotein Receptor-Related Protein-1/metabolism , Male , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/physiology , Middle Aged , RNA, Messenger/metabolism , Vascular Endothelial Growth Factor A
3.
Sci Rep ; 5: 12218, 2015 Jul 17.
Article in English | MEDLINE | ID: mdl-26183281

ABSTRACT

In vitro cultured brain tumour cells are indispensable tools for drug screening and therapeutic development. Serum-free culture conditions tentatively preserve the features of the original tumour, but commonly comprise neurosphere propagation, which is a technically challenging procedure. Here, we define a simple, non-expensive and reproducible serum-free cell culture protocol for establishment and propagation of primary paediatric brain tumour cultures as adherent monolayers. The success rates for establishment of primary cultures (including medulloblastomas, atypical rhabdoid tumour, ependymomas and astrocytomas) were 65% (11/17) and 78% (14/18) for sphere cultures and monolayers respectively. Monolayer culturing was particularly feasible for less aggressive tumour subsets, where neurosphere cultures could not be generated. We show by immunofluorescent labelling that monolayers display phenotypic similarities with corresponding sphere cultures and primary tumours, and secrete clinically relevant inflammatory factors, including PGE2, VEGF, IL-6, IL-8 and IL-15. Moreover, secretion of PGE2 was considerably reduced by treatment with the COX-2 inhibitor Valdecoxib, demonstrating the functional utility of our newly established monolayer for preclinical therapeutic assays. Our findings suggest that this culture method could increase the availability and comparability of clinically representative in vitro models of paediatric brain tumours, and encourages further molecular evaluation of serum-free monolayer cultures.


Subject(s)
Brain Neoplasms/pathology , Culture Media, Serum-Free , Drug Screening Assays, Antitumor , Primary Cell Culture , Adolescent , Antigens, CD/metabolism , Antineoplastic Agents/pharmacology , Biomarkers , Brain Neoplasms/drug therapy , Brain Neoplasms/metabolism , Cell Adhesion , Cell Proliferation/drug effects , Child , Child, Preschool , Cyclooxygenase 2/metabolism , Cytokines/metabolism , Dinoprostone/metabolism , Drug Screening Assays, Antitumor/methods , Female , Humans , Male , Neoplasm Grading , Phenotype , Primary Cell Culture/methods , Spheroids, Cellular , Tumor Cells, Cultured
4.
J Neuroimmunol ; 274(1-2): 240-3, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25086876

ABSTRACT

Bone marrow-derived mesenchymal stromal cells (MSCs) target glioma extensions and micro-satellites efficiently when implanted intratumorally. Here, we report that intratumoral implantation of MSCs and peripheral immunotherapy with interferon-gamma (IFNγ) producing tumor cells improve the survival of glioma-bearing rats (54% cure rate) compared to MSC alone (0% cure rate) or immunotherapy alone (21% cure rate) by enforcing an intratumoral CD8(+) T cell response. Further analysis revealed that the MSCs up-regulate MHC classes I and II in response to IFNγ treatment in vitro and secrete low amounts of immunosuppressive molecules prostaglandin E2 and interleukin-10.


Subject(s)
Brain Neoplasms/therapy , Glioma/therapy , Immunotherapy/methods , Mesenchymal Stem Cell Transplantation/methods , Mesenchymal Stem Cells/immunology , Animals , Brain Neoplasms/immunology , Cell Line, Tumor , Glioma/immunology , Injections, Intralesional , Male , Rats , Rats, Inbred F344 , T-Lymphocytes/immunology
5.
J Neuroimmunol ; 274(1-2): 161-7, 2014 Sep 15.
Article in English | MEDLINE | ID: mdl-25022336

ABSTRACT

Malignant brain tumors induce pronounced immunosuppression, which diminishes immune responses generated by immunotherapy. Here we report that peripheral immunotherapy, using irradiated unmodified whole tumor cells, and systemic cyclooxygenase-2 inhibition induce cure in glioma-bearing rats (60% cure rate), whereas neither monotherapy was sufficient to cure any animal. Moreover, the combined therapy protected against secondary tumor challenges (89% cure rate) and the secondary immune response was correlated with increased plasma interferon-gamma levels and CD8(+) T cells systemically and intratumorally. In conclusion, we demonstrate that cyclooxygenase-2 inhibition is sufficient to render unmodified tumor cells immunogenic in immunotherapy of experimental brain tumors.


Subject(s)
Brain Neoplasms/drug therapy , CD8-Positive T-Lymphocytes/drug effects , Cyclooxygenase 2 Inhibitors/pharmacology , Glioma/drug therapy , Immunologic Memory/drug effects , Animals , Brain Neoplasms/immunology , Brain Neoplasms/pathology , CD8-Positive T-Lymphocytes/immunology , Cell Line, Tumor , Disease Models, Animal , Glioma/immunology , Glioma/pathology , Immunization/methods , Immunologic Memory/immunology , Interferon-gamma/blood , Interferon-gamma/immunology , Male , Rats , Rats, Inbred F344
6.
Int J Cancer ; 134(11): 2748-53, 2014 Jun 01.
Article in English | MEDLINE | ID: mdl-24243648

ABSTRACT

Immunotherapy has shown effectiveness against experimental malignant brain tumors, but the clinical results have been less convincing most likely due to immunosuppression. Prostaglandin E2 (PGE2 ) is the key immunosuppressive product of cyclooxygenase-2 (COX-2) and increased levels of PGE2 and COX-2 have been shown in several tumor types, including brain tumors. In the current study, we report enhanced cure rate of mice with established mouse GL261 brain tumors when immunized with granulocyte macrophage-colony stimulating factor (GM-CSF) secreting tumor cells and simultaneously treated with the selective COX-2 inhibitors parecoxib systemically (5 mg/kg/day; 69% cure rate) or valdecoxib intratumorally (5.3 µg/kg/day; 63% cure rate). Both combined therapies induced a systemic antitumor response of proliferating CD4(+) and CD8(+) T cells, and further analysis revealed T helper 1 (Th 1) cell supremacy. The GL261 tumor cell line produced low levels of PGE2 in vitro, and co-staining at the tumor site demonstrated that a large fraction of the COX-2(+) cells were derived from CD45(+) immune cells and more specifically macrophages (F4/80(+)), indicating that tumor-infiltrating immune cells constitute the primary source of COX-2 and PGE2 in this model. We conclude that intratumoral COX-2 inhibition potentiates GM-CSF immunotherapy against established brain tumors at substantially lower doses than systemic administration. These findings underscore the central role of targeting COX-2 during immunotherapy and implicate intratumoral COX-2 as the primary target.


Subject(s)
Brain Neoplasms/therapy , Cyclooxygenase 2 Inhibitors/therapeutic use , Cyclooxygenase 2/chemistry , Glioma/therapy , Granulocyte-Macrophage Colony-Stimulating Factor/therapeutic use , Immunotherapy , Animals , Antineoplastic Combined Chemotherapy Protocols , Brain Neoplasms/immunology , Brain Neoplasms/metabolism , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation , Cyclooxygenase 2/metabolism , Dinoprostone/metabolism , Drug Synergism , Female , Flow Cytometry , Glioma/immunology , Glioma/metabolism , Immunoenzyme Techniques , Intramolecular Oxidoreductases/metabolism , Isoxazoles/therapeutic use , Mice , Mice, Inbred C57BL , Prostaglandin-E Synthases , Sulfonamides/therapeutic use , Tumor Cells, Cultured
7.
Cancer Immunol Immunother ; 62(9): 1463-74, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23775421

ABSTRACT

Despite temozolomide (TMZ) treatment, the prognosis for patients with glioblastoma multiforme is still dismal. As dose escalation of TMZ is limited by systemic toxicity, intratumoral delivery emerges as an attractive treatment modality, which may sustain cytotoxic drug concentrations intratumorally and induce immunogenic cell death. Both clinical and experimental gliomas have responded to immunotherapy, but the benefit of simultaneous chemo- and immunotherapy is inadequately studied. Here, we monitored survival of GL261-bearing C57BL/6 mice following a 3-day treatment with either intratumoral TMZ (micro-osmotic pump, 4.2 mg/kg/day) or systemic TMZ (i.p. injections, 50 mg/kg/day) alone, or combined with immunization using GM-CSF secreting GL261 cells. Peripheral and intratumoral leukocytes were analyzed by flow cytometry and immunohistochemistry. Intratumoral TMZ induced higher survival rate than systemic TMZ (45 vs. 8%). When T cells were depleted following intratumoral TMZ, the therapeutic effect was completely abrogated (0 % survival). Intratumoral TMZ synergistically increased survival rate of immunized mice (from 25 to 83%), while systemic TMZ failed (0%). While systemic TMZ induced a transient leukopenia, intratumoral TMZ and immunotherapy sustained the proliferation of CD8+ T cells and decreased the number of intratumoral immunosuppressive cells. In conclusion, intratumoral TMZ alone or in combination with immunotherapy could cure glioma-bearing mice, due to attenuation of local immunosuppression and increase in potential effector immune cells.


Subject(s)
Antineoplastic Agents, Alkylating/administration & dosage , Dacarbazine/analogs & derivatives , Glioma/therapy , Immunotherapy, Adoptive/methods , Animals , Cell Line, Tumor , Combined Modality Therapy , Dacarbazine/administration & dosage , Female , Glioma/drug therapy , Glioma/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/immunology , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Humans , Injections, Intralesional , Mice , Mice, Inbred C57BL , Temozolomide
8.
J Neuroimmunol ; 258(1-2): 91-5, 2013 May 15.
Article in English | MEDLINE | ID: mdl-23528658

ABSTRACT

Peripheral immunization, using a combination of interferon-gamma (IFNγ)- and interleukin-7 (IL-7)-producing tumor cells, eradicated 75% of pre-established intracerebral N32 rat glioma tumors, and prolonged survival in the more aggressive RG2 model. Rats immunized with IFNγ- and IL7-transduced N32 cells displayed increases in IFNγ plasma levels and proliferating circulating T cells when compared with rats immunized with N32-wild type cells. Following irradiation, the expression of MHC I and II was high on N32-IFNγ cells, but low on RG2-IFNγ cells. In conclusion, IFNγ and IL-7 immunizations prolong survival in two rat glioma models.


Subject(s)
Brain Neoplasms/immunology , Glioma/immunology , Immunotherapy/methods , Interferon-gamma/immunology , Interleukin-7/immunology , T-Lymphocytes/immunology , Animals , Disease Models, Animal , Interferon-gamma/administration & dosage , Interleukin-7/administration & dosage , Rats
9.
Cancer Immunol Immunother ; 61(8): 1191-9, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22213142

ABSTRACT

Glioblastoma multiforme is the most common and aggressive malignant brain tumor in humans, and the prognosis is very poor despite conventional therapy. Immunotherapy represents a novel treatment approach, but the effect is often weakened by release of immune-suppressive molecules such as prostaglandins. In the current study, we investigated the effect of immunotherapy with irradiated interferon-γ (IFN-γ)-secreting tumor cells and administration of the selective cyclooxygenase-2 (COX-2) inhibitor parecoxib as treatment of established rat brain tumors. COX-2 inhibition and immunotherapy significantly enhanced the long-term cure rate (81% survival) compared with immunotherapy alone (19% survival), and there was a significant increase in plasma IFN-γ levels in animals treated with the combined therapy, suggesting a systemic T helper 1 immune response. COX-2 inhibition alone, however, did neither induce cure nor prolonged survival. The tumor cells were identified as the major source of COX-2 both in vivo and in vitro, and unmodified tumor cells produced prostaglandin E(2) in vitro, while the IFN-γ expressing tumor cells secreted significantly lower levels. In conclusion, we show that immunotherapy of experimental brain tumors is greatly potentiated when combined with COX-2 inhibition. Based on our results, the clinically available drug parecoxib may be added to immunotherapy against human brain tumors. Furthermore, the discovery that IFN-γ plasma levels can be used to determine the ongoing in vivo immune response has translational potential.


Subject(s)
Cyclooxygenase 2 Inhibitors/pharmacology , Cyclooxygenase 2/metabolism , Glioblastoma/therapy , Immunotherapy, Adoptive/methods , Neoplasms, Experimental/enzymology , Neoplasms, Experimental/therapy , Animals , Brain Neoplasms/enzymology , Brain Neoplasms/immunology , Brain Neoplasms/therapy , Combined Modality Therapy , Disease Models, Animal , Flow Cytometry , Glioblastoma/enzymology , Glioblastoma/immunology , Immunohistochemistry , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Isoxazoles/pharmacology , Male , Neoplasms, Experimental/immunology , Rats , Rats, Inbred F344
10.
Int J Cancer ; 124(3): 630-7, 2009 Feb 01.
Article in English | MEDLINE | ID: mdl-18972433

ABSTRACT

We were the first to demonstrate that combined immunotherapy with GM-CSF producing GL261 cells and recombinant IFNgamma of preestablished GL261 gliomas could cure 90% of immunized mice. To extend these findings and to uncover the underlying mechanisms, the ensuing experiments were undertaken. We hypothesized that immunizations combining both GM-CSF and IFNgamma systemically would increase the number of immature myeloid cells, which then would mature and differentiate into dendritic cells (DCs) and macrophages, thereby augmenting tumor antigen presentation and T-cell activation. Indeed, the combined therapy induced a systemic increase of both immature and mature myeloid cells but also an increase in T regulatory cells (T-regs). Cytotoxic anti-tumor responses, mirrored by an increase in Granzyme B-positive cells as well as IFNgamma-producing T-cells, were augmented after immunizations with GM-CSF and IFNgamma. We also show that the combined therapy induced a long-term memory with rejection of intracerebral (i.c.) rechallenges. Depletion of T-cells showed that both CD4+ and CD8+ T-cells were essential for the combined GM-CSF and IFNgamma effect. Finally, when immunizations were delayed until day 5 after tumor inoculation, only mice receiving immunotherapy with both GM-CSF and IFNgamma survived. We conclude that the addition of recombinant IFNgamma to immunizations with GM-CSF producing tumor cells increased the number of activated tumoricidal T-cells, which could eradicate established intracerebral tumors. These results clearly demonstrate that the combination of cytokines in immunotherapy of brain tumors have synergistic effects that have implications for clinical immunotherapy of human malignant brain tumors.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Glioma/drug therapy , Glioma/immunology , Immunotherapy/methods , Animals , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Flow Cytometry , Granulocyte-Macrophage Colony-Stimulating Factor/administration & dosage , Immunologic Memory/drug effects , Interferon-gamma/administration & dosage , Male , Mice , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL
...