Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 9 de 9
Filter
Add more filters










Database
Language
Publication year range
1.
Cancers (Basel) ; 13(2)2021 Jan 19.
Article in English | MEDLINE | ID: mdl-33478100

ABSTRACT

(1) Background: Despite the indisputable effectiveness of dexamethasone (DEXA) to reduce inflammation in glioblastoma (GBM) patients, its influence on tumour progression and radiotherapy response remains controversial. (2) Methods: We analysed patient data and used expression and cell biological analyses to assess effects of DEXA on GBM cells. We tested the efficacy of tyrosine kinase inhibitors in vitro and in vivo. (3) Results: We confirm in our patient cohort that administration of DEXA correlates with worse overall survival and shorter time to relapse. In GBM cells and glioma stem-like cells (GSCs) DEXA down-regulates genes controlling G2/M and mitotic-spindle checkpoints, and it enables cells to override the spindle assembly checkpoint (SAC). Concurrently, DEXA up-regulates Platelet Derived Growth Factor Receptor (PDGFR) signalling, which stimulates expression of anti-apoptotic regulators BCL2L1 and MCL1, required for survival during extended mitosis. Importantly, the protective potential of DEXA is dependent on intact tyrosine kinase signalling and ponatinib, sunitinib and dasatinib, all effectively overcome the radio-protective and pro-proliferative activity of DEXA. Moreover, we discovered that DEXA-induced signalling creates a therapeutic vulnerability for sunitinib in GSCs and GBM cells in vitro and in vivo. (4) Conclusions: Our results reveal a novel DEXA-induced mechanism in GBM cells and provide a rationale for revisiting the use of tyrosine kinase inhibitors for the treatment of GBM.

2.
Pancreatology ; 21(1): 215-223, 2021 Jan.
Article in English | MEDLINE | ID: mdl-33358592

ABSTRACT

BACKGROUND: Pancreatic neuroendocrine tumors are rare neoplasms for which few predictive and/or prognostic biomarkers have been validated. Our previous work suggested the potential of the combined expression of N-myc downstream-regulated gen-1 (NDRG-1), O6-methylguanine DNA methyltransferase (MGMT) and Pleckstrin homology-like domain family A member 3 (PHLDA-3) as prognostic factors for relapse and survival. METHODS: In this new multicenter study we evaluated immunohistochemistry expression in 76 patients with advanced PanNET who were treated with capecitabine-temozolomide or everolimus. Based on the immunohistochemistry panel, an immunohistochemistry prognostic score (IPS) was developed. RESULTS: In patients treated with capecitabine and temozolomide, low IPS was an independent prognostic factor for progression-free-survival and overall-survival. Similar findings were observed with highest IPS for overall-survival in patients treated with everolimus. CONCLUSION: From our knowledge, it is the first time that a simple IPS could be useful to predict outcome for patients with metastatic pancreatic neuroendocrine tumors treated with everolimus or capecitabine and temozolomide.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Everolimus/therapeutic use , Immunohistochemistry/methods , Immunosuppressive Agents/therapeutic use , Neuroendocrine Tumors/drug therapy , Neuroendocrine Tumors/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/analysis , Cell Cycle Proteins/analysis , Cell Line, Tumor , DNA Modification Methylases/analysis , DNA Repair Enzymes/analysis , Female , Humans , Intracellular Signaling Peptides and Proteins/analysis , Male , Middle Aged , Neoplasm Recurrence, Local , Neuroendocrine Tumors/mortality , Nuclear Proteins/analysis , Pancreatic Neoplasms/mortality , Prognosis , Progression-Free Survival , Survival Analysis , Tumor Suppressor Proteins/analysis , Young Adult
3.
J Immunol ; 195(7): 3180-9, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26324768

ABSTRACT

Regulatory T cell (Treg) activity is modulated by a cooperative complex between the transcription factor NFAT and FOXP3, a lineage specification factor for Tregs. FOXP3/NFAT interaction is required to repress expression of IL-2, upregulate expression of the Treg markers CTLA4 and CD25, and confer suppressor function to Tregs. However, FOXP3 is expressed transiently in conventional CD4(+) T cells upon TCR stimulation and may lead to T cell hyporesponsiveness. We found that a short synthetic peptide able to inhibit FOXP3/NFAT interaction impaired suppressor activity of conventional Tregs in vitro. Specific inhibition of FOXP3/NFAT interaction with this inhibitory peptide revealed that FOXP3 downregulates NFAT-driven promoter activity of CD40L and IL-17. Inhibition of FOXP3/NFAT interaction upregulated CD40L expression on effector T cells and enhanced T cell proliferation and IL-2, IFN-γ, IL-6, or IL-17 production in response to TCR stimulation. The inhibitory peptide impaired effector T cell conversion into induced Tregs in the presence of TGF-ß. Moreover, in vivo peptide administration showed antitumor efficacy in mice bearing Hepa129 or TC1 tumor cells when combined with sorafenib or with an antitumor vaccine, respectively. Our results suggest that inhibition of NFAT/FOXP3 interaction might improve antitumor immunotherapies.


Subject(s)
CD40 Ligand/biosynthesis , Forkhead Transcription Factors/metabolism , Interleukin-17/biosynthesis , NFATC Transcription Factors/metabolism , Receptors, Antigen, T-Cell/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antineoplastic Agents/pharmacology , CD40 Ligand/genetics , CTLA-4 Antigen/biosynthesis , Cell Proliferation/genetics , Female , Forkhead Transcription Factors/antagonists & inhibitors , Humans , Immunotherapy , Interferon-gamma/biosynthesis , Interleukin-17/genetics , Interleukin-2/biosynthesis , Interleukin-2 Receptor alpha Subunit/biosynthesis , Interleukin-6/biosynthesis , Jurkat Cells , Lymphocyte Activation/immunology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , NFATC Transcription Factors/antagonists & inhibitors , Neoplasms/therapy , Niacinamide/analogs & derivatives , Niacinamide/pharmacology , Ovalbumin/immunology , Peptide Fragments/pharmacology , Phenylurea Compounds/pharmacology , Promoter Regions, Genetic/genetics , Sorafenib , Transforming Growth Factor beta/metabolism
4.
Biomed Res Int ; 2013: 864720, 2013.
Article in English | MEDLINE | ID: mdl-24093105

ABSTRACT

The development of tools for efficient targeting of antigens to antigen presenting cells is of great importance for vaccine development. We have previously shown that fusion proteins containing antigens fused to the extra domain A from fibronectin (EDA), an endogenous TLR4 ligand, which targets antigens to TLR4-expressing dendritic cells (DC), are highly immunogenic. To facilitate the procedure of joining EDA to any antigen of choice, we have prepared the fusion protein EDAvidin by linking EDA to the N terminus of streptavidin, allowing its conjugation with biotinylated antigens. We found that EDAvidin, as streptavidin, forms tetramers and binds biotin or biotinylated proteins with a Kd ~ 2.6 × 10(-14) mol/L. EDAvidin favours the uptake of biotinylated green fluorescent protein by DC. Moreover, EDAvidin retains the proinflammatory properties of EDA, inducing NF- κß by TLR4-expressing cells, as well as the production of TNF- α by the human monocyte cell line THP1 and IL-12 by DC. More importantly, immunization of mice with EDAvidin conjugated with the biotinylated nonstructural NS3 protein from hepatitis C virus induces a strong anti-NS3 T cell immune response. These results open a new way to use the EDA-based delivery tool to target any antigen of choice to DC for vaccination against infectious diseases and cancer.


Subject(s)
Antigen-Presenting Cells/immunology , Fibronectins/immunology , Recombinant Fusion Proteins/immunology , Toll-Like Receptor 4/immunology , Animals , Antigen-Presenting Cells/metabolism , Cell Line , Dendritic Cells/immunology , Dendritic Cells/metabolism , Fibronectins/chemistry , Fibronectins/genetics , Hepacivirus/immunology , Humans , Immunotherapy, Active , Ligands , Mice , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Streptavidin/chemistry , T-Lymphocytes/immunology , T-Lymphocytes/pathology , Toll-Like Receptor 4/metabolism
5.
Vaccine ; 30(18): 2848-58, 2012 Apr 16.
Article in English | MEDLINE | ID: mdl-22387222

ABSTRACT

The complement system and Toll-like receptors (TLR) are key innate defense systems which might interact synergistically on dendritic cells (DC) to reinforce adaptive immunity. In a previous work, we found that the extra domain A from fibronectin EDA (an endogenous ligand for TLR4) can favour antigen delivery to DC and induce their maturation. Given the potential of anaphylatoxins to cause inflammation and activation of myeloid cells, we hypothesized that a fusion protein between EDA, and anaphylatoxins C3a, C4a or C5a together with an antigen might improve the immunogenicity of the antigen. Naked DNA immunization with a construct expressing the fusion protein between C5a, EDA and the cytotoxic T cell epitope SIINFEKL from ovalbumin, induced strong antigen specific T cell responses. The purified recombinant fusion protein EDA-SIINFEKL-C5a induced activation of dendritic cells, the production of proinflammatory cytokines/chemokines and stimulated antigen presenting cell migration and NK cell activation. As compared to EDA-SIINFEKL, the fusion protein EDA-SIINFEKL-C5a did not induce the production of the immunosuppressive molecules IL-10, CCL17, CCL1, CXCL12 or XCL1 by DC. Moreover, EDA-SIINFEKL-C5a induced strong specific T cell responses in vivo and protected mice against E.G7-OVA tumor growth more efficiently than EDA-SIINFEKL or SIINFEKL-C5a recombinant proteins. Our results suggest that fusion proteins containing EDA, the anaphylatoxin C5a and the antigen may serve as a suitable strategy for the development of anti-tumor or anti-viral vaccines.


Subject(s)
Anaphylatoxins/immunology , Complement C5a/immunology , Ectodysplasins/immunology , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/agonists , Anaphylatoxins/genetics , Animals , Cancer Vaccines/administration & dosage , Cancer Vaccines/immunology , Complement C5a/genetics , Cytokines/metabolism , Ectodysplasins/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/immunology , Female , Mice , Mice, Inbred C57BL , Neoplasms/immunology , Neoplasms/prevention & control , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/immunology , Vaccines, DNA/administration & dosage , Vaccines, DNA/immunology
6.
Int J Cancer ; 131(3): 641-51, 2012 Aug 01.
Article in English | MEDLINE | ID: mdl-21898393

ABSTRACT

Cervical carcinoma is one of the most common cancers in women worldwide. It is well established that chronic infection of the genital tract by various mucosatropic human papillomavirus (HPV) types causes cervical cancer. Cellular immunity to E7 protein from HPV (HPVE7) has been associated with clinical and cytologic resolution of HPV-induced lesions. Thus, we decided to test if targeting of HPVE7 to dendritic cells using a fusion protein containing the extra domain A (EDA) from fibronectin, a natural ligand for TLR4, and HPVE7 (EDA-HPVE7) might be an efficient vaccine for the treatment of cervical carcinoma. We found that EDA-HPVE7 fusion protein was efficiently captured by bone marrow derived dendritic cells in vitro and induced their maturation, with the upregulation of maturation markers and the production of IL-12. Immunization of mice with EDA-HPVE7 fusion protein induced antitumor CD8(+) T cell responses in the absence of additional adjuvants. Repeated intratumoral administration of EDA-HPVE7 in saline was able to cure established TC-1 tumors of 5-7 mm in diameter. More importantly, intravenous injection with EDA-HPVE7 in combination with the TLR ligand polyinosinic-polycytidylic acid (pIC), or with low doses of cyclophosphamide and the TLR9 ligand CpG-B complexed in cationic lipids, were able to eradicate large established TC-1 tumors (1.2 cm in diameter). Thus, therapeutic vaccination with EDA-HPVE7 fusion protein may be effective in the treatment of human cervical carcinoma.


Subject(s)
Cancer Vaccines/immunology , Fibronectins/immunology , Papillomavirus E7 Proteins/immunology , Papillomavirus Vaccines/immunology , Uterine Cervical Neoplasms/immunology , Uterine Cervical Neoplasms/therapy , Adjuvants, Immunologic , Animals , Bone Marrow Cells/immunology , CD8-Positive T-Lymphocytes/immunology , Cyclophosphamide/pharmacology , Dendritic Cells/immunology , Female , Fibronectins/metabolism , HEK293 Cells , Humans , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology , Papillomaviridae/immunology , Papillomavirus E7 Proteins/metabolism , Papillomavirus Infections/immunology , Poly I-C/pharmacology , Recombinant Fusion Proteins/immunology , Uterine Cervical Neoplasms/virology , Vaccines, Synthetic/immunology
7.
Cancer Res ; 71(9): 3214-24, 2011 May 01.
Article in English | MEDLINE | ID: mdl-21402711

ABSTRACT

Low antigen expression and an absence of coimmunostimulatory signals may be partly responsible for the low immunogenicity of many tumors. It may be possible to overcome this situation by defining a combination of adjuvants and antigens that can activate a high-avidity antitumor response. Using the poorly immunogenic B16-OVA melanoma cells as tumor model, we tested different combinations of adjuvants and antigens to treat established tumors. In the absence of exogenous antigens, repeated administration of the TLR7 ligand Imiquimod together with anti-CD40 agonistic antibodies activated only innate immunity, which was insufficient to reject intradermal tumors. Administering this adjuvant combination together with OVA as a tumor antigen induced T-cell responses that delayed tumor growth. However, administering a combination of anti-CD40 plus TLR3 and TLR7 ligands, together with antigen targeting to dendritic cells through TLR4, was sufficient to induce tumor rejection in 50% of mice. This response was associated with a greater activation of innate immunity and induction of high-avidity polyfunctional CD8(+) T-cell responses, which each contributed to tumor rejection. This therapy activated T-cell responses not only against OVA, which conferred protection against a rechallenge with B16-OVA cells, but also activated T-cell responses against other melanoma-associated antigens. Our findings support the concept that multiple adjuvant combination and antigen targeting may be a useful immunotherapeutic strategy against poorly immunogenic tumors.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigens, Neoplasm/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/therapy , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Aminoquinolines/pharmacology , Animals , Antibodies/immunology , Antibodies/pharmacology , Antibody Affinity , Antigens, Neoplasm/pharmacology , CD4 Antigens/immunology , Female , Imiquimod , Immunity, Innate/drug effects , Mice , Mice, Inbred C57BL , Ovalbumin/genetics , Ovalbumin/immunology , Ovalbumin/pharmacology , Poly I-C/pharmacology , Thymoma/genetics , Thymoma/immunology , Transfection
8.
Vaccine ; 28(44): 7146-54, 2010 Oct 18.
Article in English | MEDLINE | ID: mdl-20817012

ABSTRACT

Staphylococcus epidermidis releases a complex of at least four peptides, termed phenol-soluble modulins (PSM), which stimulate macrophages to produce proinflammatory cytokines via activation of TLR2 signalling pathway. We demonstrated that covalent linkage of PSM peptides to an antigen facilitate its capture by dendritic cells and, in combination with different TLR ligands, can favour the in vivo induction of strong and persistent antigen-specific immune responses. Treatment of mice grafted with HPV16-E7-expressing tumor cells (TC-1) with poly(I:C) and a peptide containing αMod linked to the H-2D(b)-restricted cytotoxic T-cell epitope E7(49-57) from HPV16-E7 protein allowed complete tumor regression in 100% of the animals. Surprisingly, this immunomodulatory property of modulin-derived peptides was TLR2 independent and partially dependent upon the EGF-receptor signalling pathway. Our results suggest that alpha or gamma modulin peptides may serve as a suitable antigen carrier for the development of anti-tumoral or anti-viral vaccines.


Subject(s)
Antigens, Neoplasm/immunology , Bacterial Toxins/immunology , Neoplasms, Experimental/drug therapy , Papillomavirus E7 Proteins/immunology , Animals , Antigen Presentation , Antigens, Neoplasm/administration & dosage , Bacterial Toxins/administration & dosage , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/immunology , ErbB Receptors/metabolism , Female , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms, Experimental/prevention & control , Papillomavirus E7 Proteins/administration & dosage , Poly I-C/immunology , Staphylococcus epidermidis , Toll-Like Receptor 2/immunology
9.
J Immunol ; 178(2): 748-56, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17202335

ABSTRACT

Vaccination strategies based on the in vivo targeting of Ags to dendritic cells (DCs) are needed to improve the induction of specific T cell immunity against tumors and infectious agents. In this study, we have used a recombinant protein encompassing the extra domain A from fibronectin (EDA), an endogenous ligand for TLR4, to deliver Ags to TLR4-expressing DC. The purified EDA protein was shown to bind to TLR4-expressing HEK293 cells and to activate the TLR4 signaling pathway. EDA also stimulated the production by DC of proinflammatory cytokines such as IL-12 or TNF-alpha and induced their maturation in vitro and in vivo. A fusion protein between EDA and a cytotoxic T cell epitope from OVA efficiently presented this epitope to specific T cells and induced the in vivo activation of a strong and specific CTL response. Moreover, a fusion protein containing EDA and the full OVA also improved OVA presentation by DC and induced CTL responses in vivo. These EDA recombinant proteins protected mice from a challenge with tumor cells expressing OVA. These results strongly suggest that the fibronectin extra domain A may serve as a suitable Ag carrier for the development of antiviral or antitumoral vaccines.


Subject(s)
Fibronectins/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 4/metabolism , Amino Acid Motifs , Animals , Bone Marrow/metabolism , Cell Differentiation , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Female , Fibronectins/chemistry , Fibronectins/genetics , Humans , Interleukin-12/biosynthesis , Mice , Mice, Inbred C57BL , Mice, Knockout , Neoplasms/immunology , Neoplasms/metabolism , Neoplasms/pathology , Ovalbumin/immunology , Peptide Fragments/chemistry , Peptide Fragments/genetics , Peptide Fragments/metabolism , Protein Binding , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Signal Transduction , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics , Tumor Necrosis Factor-alpha/biosynthesis
SELECTION OF CITATIONS
SEARCH DETAIL