Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Oncoimmunology ; 5(2): e1075113, 2016 Feb.
Article in English | MEDLINE | ID: mdl-27057445

ABSTRACT

Blocking antibodies against immunosuppressive molecules have shown promising results in cancer patients. However, there are not enough data to define those conditions dictating treatment efficacy. In this scenario, IL-10 is a cytokine with controversial effects on tumor growth. Thus, our aim was to characterize in which setting IL-10 blockade may potentiate the beneficial effects of a therapeutic vaccine In the IL-10-expressing B16-OVA and TC-1 P3 (A15) tumor models, therapeutic vaccination with tumor antigens plus the TLR7 ligand Imiquimod increased IL-10 production. Although blockade of IL-10 signal with anti-IL-10R antibodies did not inhibit tumor growth, when combined with vaccination it enhanced tumor rejection, associated with stronger innate and adaptive immune responses. Interestingly, a similar enhancement on immune responses was observed after simultaneous vaccination and IL-10 blockade in naive mice. However, when using vaccines containing as adjuvants the TLR3 ligand poly(I:C) or anti-CD40 agonistic antibodies, despite tumor IL-10 expression, anti-IL-10R antibodies did not provide any beneficial effect on tumor growth and antitumor immune responses. Of note, as opposed to Imiquimod, vaccination with this type of adjuvants did not induce IL-10 and correlated with a lack of in vitro IL-10 production by dendritic cells (DC). Finally, in B16-OVA-bearing mice, blockade of IL-10 during therapeutic vaccination with a multiple adjuvant combination (MAC) with potent immunostimulatory properties but still inducing IL-10 led to superior antitumor immunity and complete tumor rejection. These results suggest that for therapeutic antitumor vaccination, blockade of vaccine-induced IL-10 is more relevant than tumor-associated IL-10.

2.
Blood ; 125(20): 3153-63, 2015 May 14.
Article in English | MEDLINE | ID: mdl-25977583

ABSTRACT

Tumor-mediated procoagulatory activity leads to venous thromboembolism and supports metastasis in cancer patients. A prerequisite for metastasis formation is the interaction of cancer cells with endothelial cells (ECs) followed by their extravasation. Although it is known that activation of ECs and the release of the procoagulatory protein von Willebrand factor (VWF) is essential for malignancy, the underlying mechanisms remain poorly understood. We hypothesized that VWF fibers in tumor vessels promote tumor-associated thromboembolism and metastasis. Using in vitro settings, mouse models, and human tumor samples, we showed that melanoma cells activate ECs followed by the luminal release of VWF fibers and platelet aggregation in tumor microvessels. Analysis of human blood samples and tumor tissue revealed that a promoted VWF release combined with a local inhibition of proteolytic activity and protein expression of ADAMTS13 (a disintegrin-like and metalloproteinase with thrombospondin type I repeats 13) accounts for this procoagulatory milieu. Blocking endothelial cell activation by the low-molecular-weight heparin tinzaparin was accompanied by a lack of VWF networks and inhibited tumor progression in a transgenic mouse model. Our findings implicate a mechanism wherein tumor-derived vascular endothelial growth factor-A (VEGF-A) promotes tumor progression and angiogenesis. Thus, targeting EC activation envisions new therapeutic strategies attenuating tumor-related angiogenesis and coagulation.


Subject(s)
Melanoma/metabolism , Platelet Aggregation , von Willebrand Factor/metabolism , ADAM Proteins/blood , ADAM Proteins/metabolism , ADAMTS13 Protein , Animals , Blood Coagulation , Blood Platelets , Disease Models, Animal , Disease Progression , Endothelial Cells/metabolism , Enzyme Activation , Fibrinolytic Agents/pharmacology , Heparin, Low-Molecular-Weight/pharmacology , Humans , Melanoma/blood , Melanoma/pathology , Mice , Mice, Transgenic , Microvessels/metabolism , Neovascularization, Pathologic/metabolism , Protein Binding , Proto-Oncogene Proteins c-ret/metabolism , Tinzaparin , Vascular Endothelial Growth Factor A/antagonists & inhibitors , Vascular Endothelial Growth Factor A/metabolism
3.
PLoS One ; 9(5): e96799, 2014.
Article in English | MEDLINE | ID: mdl-24797128

ABSTRACT

Transforming growth factor ß (TGF-ß) is a powerful promoter of cancer progression and a key target for antitumor therapy. As cancer cells exhibit active cholesterol metabolism, high density lipoproteins (HDLs) appear as an attractive delivery system for anticancer TGFß-inhibitory molecules. We constructed a plasmid encoding a potent TGF-ß-blocking peptide (P144) linked to apolipoprotein A-I (ApoA-I) through a flexible linker (pApoLinkerP144). The ApoLinkerP144 sequence was then incorporated into a hepatotropic adeno-associated vector (AAVApoLinkerP144). The aim was to induce hepatocytes to produce HDLs containing a modified ApoA-I capable of blocking TGF-ß. We observed that transduction of the murine liver with pApoLinkerP144 led to the appearance of a fraction of circulating HDL containing the fusion protein. These HDLs were able to attenuate TGF-ß signaling in the liver and to enhance IL-12 -mediated IFN-γ production. Treatment of liver metastasis of MC38 colorectal cancer with AAVApoLinkerP144 resulted in a significant reduction of tumor growth and enhanced expression of IFN-γ and GM-CSF in cancerous tissue. ApoLinkerP144 also delayed MC38 liver metastasis in Rag2-/-IL2rγ-/- immunodeficient mice. This effect was associated with downregulation of TGF-ß target genes essential for metastatic niche conditioning. Finally, in a subset of ret transgenic mice, a model of aggressive spontaneous metastatic melanoma, AAVApoLinkerP144 delayed tumor growth in association with increased CD8+ T cell numbers in regional lymph nodes. In conclusion, modification of HDLs to transport TGF-ß-blocking molecules is a novel and promising approach to inhibit the growth of liver metastases by immunological and non-immunological mechanisms.


Subject(s)
Colorectal Neoplasms/pathology , Lipoproteins, HDL/pharmacology , Liver Neoplasms/metabolism , Liver Neoplasms/pathology , Molecular Targeted Therapy , Transforming Growth Factor beta1/metabolism , Animals , CD3 Complex/metabolism , Colorectal Neoplasms/drug therapy , Colorectal Neoplasms/metabolism , Female , Genetic Vectors , Granulocyte-Macrophage Colony-Stimulating Factor/metabolism , Hepatocytes/metabolism , Interferon-gamma/metabolism , Liver/metabolism , Liver Neoplasms/drug therapy , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Transgenic , Neoplasm Metastasis/prevention & control , Plasmids/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins c-ret/genetics , Transforming Growth Factor beta1/antagonists & inhibitors
4.
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
5.
Cancer Res ; 71(3): 812-21, 2011 Feb 01.
Article in English | MEDLINE | ID: mdl-21159663

ABSTRACT

Melanoma progression is associated with the expression of different growth factors, cytokines, and chemokines. Because TGFß1 is a pleiotropic cytokine involved not only in physiologic processes but also in cancer development, we analyzed in A375 human melanoma cells, the effect of TGFß1 on monocyte chemoattractant protein-1 (MCP-1) and interleukin-10 (IL-10) expression, two known factors responsible for melanoma progression. TGFß1 increased the expression of MCP-1 and IL-10 in A375 cells, an effect mediated by the cross-talk between Smad, PI3K (phosphoinositide 3-kinase)/AKT, and BRAF-MAPK (mitogen activated protein kinase) signaling pathways. Supernatants from TGFß1-treated A375 cells enhanced MCP-1-dependent migration of monocytes, which, in turn, expressed high levels of TGF,ß1, bFGF, and VEGF mRNA. Moreover, these supernatants also inhibited functional properties of dendritic cells through IL-10-dependent mechanisms. When using in vitro, the TGFß1-blocking peptide P144, TGFß1-dependent Smad3 phosphorylation, and expression of MCP-1 and IL-10 were inhibited. In vivo, treatment of A375 tumor-bearing athymic mice with P144 significantly reduced tumor growth, associated with a lower macrophage infiltrate and decreased intratumor MCP-1 and VEGF levels, as well as angiogenesis. Finally, in C57BL/6 mice with B16-OVA melanoma tumors, when administered with immunotherapy, P144 decreased tumor growth and intratumor IL-10 levels, linked to enhanced activation of dendritic cells and natural killer cells, as well as anti-OVA T-cell responses. These results show new effects of TGFß1 on melanoma cells, which promote tumor progression and immunosuppression, strongly reinforcing the relevance of this cytokine as a molecular target in melanoma.


Subject(s)
Chemokine CCL2/biosynthesis , Interleukin-10/biosynthesis , Melanoma/immunology , Transforming Growth Factor beta1/pharmacology , Amino Acid Sequence , Animals , Female , Humans , Interleukin-10/immunology , MAP Kinase Signaling System , Melanoma, Experimental/immunology , Mice , Mice, Inbred C57BL , Mice, Nude , Molecular Sequence Data , Monocytes/drug effects , Monocytes/immunology , Proto-Oncogene Proteins c-akt/metabolism , Smad3 Protein/metabolism
6.
Hepatology ; 53(1): 23-31, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21154952

ABSTRACT

UNLABELLED: The high levels of interleukin 10 (IL-10) present in chronic hepatitis C virus (HCV) infection have been suggested as responsible for the poor antiviral cellular immune responses found in these patients. To overcome the immunosuppressive effect of IL-10 on antigen-presenting cells such as dendritic cells (DCs), we developed peptide inhibitors of IL-10 to restore DC functions and concomitantly induce efficient antiviral immune responses. Two IL-10-binding peptides (p9 and p13) were selected using a phage-displayed library and their capacity to inhibit IL-10 was assessed in a bioassay and in STAT-3 (signal transducer and activator of transcription 3) phosphorylation experiments in vitro. In cultures of human leukocytes where HCV core protein induces the production of IL-10, p13 restored the ability of plasmacytoid DC to produce interferon alpha (IFN-α) after Toll-like receptor 9 (TLR9) stimulation. Similarly, when myeloid DCs were stimulated with CD40L in the presence of HCV core, p9 enhanced IL-12 production by inhibiting HCV core-induced as well as CD40L-induced IL-10. Moreover, in vitro, p13 potentiated the effect of maturation stimuli on human and murine DC, increasing their IL-12 production and stimulatory activity, which resulted in enhanced proliferation and IFN-γ production by responding T-cells. Finally, immunization with p13-treated murine DC induced stronger anti-HCV T-cell responses not only in wildtype mice but also in HCV transgenic mice and in mice transiently expressing HCV core in the liver. CONCLUSION: These results suggest that IL-10 inhibiting peptides may have important applications to enhance anti-HCV immune responses by restoring the immunostimulatory capabilities of DC.


Subject(s)
Dendritic Cells/immunology , Hepacivirus/immunology , Interleukin-10/antagonists & inhibitors , Interleukin-12/biosynthesis , Amino Acid Sequence , Animals , CD40 Ligand/pharmacology , Cell Line , Dendritic Cells/metabolism , Hepatitis C Antigens/pharmacology , Humans , Interferon-alpha/biosynthesis , Interleukin-10/immunology , Mice , Peptide Library , STAT3 Transcription Factor/metabolism , Toll-Like Receptor 9/physiology , Viral Core Proteins/pharmacology
7.
J Nucleic Acids ; 20102010 Sep 23.
Article in English | MEDLINE | ID: mdl-20936147

ABSTRACT

The D. melanogaster mus308 gene, highly conserved among higher eukaryotes, is implicated in the repair of cross-links and of O-ethylpyrimidine DNA damage, working in a DNA damage tolerance mechanism. However, despite its relevance, its possible role on the processing of different DNA ethylation damages is not clear. To obtain data on mutation frequency and on mutation spectra in mus308 deficient (mus308(-)) conditions, the ethylating agent diethyl sulfate (DES) was analysed in postmeiotic male germ cells. These data were compared with those corresponding to mus308 efficient conditions. Our results indicate that Mus308 is necessary for the processing of oxygen and N-ethylation damage, for the survival of fertilized eggs depending on the level of induced DNA damage, and for an influence of the DNA damage neighbouring sequence. These results support the role of mus308 in a tolerance mechanism linked to a translesion synthesis pathway and also to the alternative end-joinig system.

8.
Int J Cancer ; 125(11): 2614-23, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19530254

ABSTRACT

Transforming growth factor-beta (TGF-beta) is a cytokine with potent immunosuppressive effects and is overexpressed in many tumors. Therefore, development of molecules able to inhibit TGF-beta is of paramount importance to improve the efficacy of antitumor immunotherapy. TGF-beta inhibitor peptides P144 and P17 were combined with the administration of adjuvant molecules poly(I:C) and agonistic anti-CD40 antibodies, and their effect on the growth of E.G7-OVA established tumors and on antitumor immune response was evaluated. Tumor rejection efficacy of a single administration of adjuvants was enhanced from 15 to 70 % when combined with repeated injections of TGF-beta inhibitor peptides. Simultaneous administration of adjuvants and TGF-beta inhibitor peptides was required for maximal therapeutic efficacy. Although tumor cells produced TGF-beta, it was found that the beneficial effect of peptide administration was mainly due to the inhibition of TGF-beta produced by regulatory CD4(+)CD25(+) T cells rather than by tumor cells. The enhanced antitumor effect was accompanied by a higher activity of dendritic cells, natural killer cells and tumor antigen-specific T cells, as well as by a decrease in the number of myeloid-derived suppressor cells. In conclusion, administration of peptide inhibitors of TGF-beta in therapeutic vaccination enhances the efficacy of immunotherapy by increasing antitumor immune responses. These peptide inhibitors may have important applications for current immunotherapeutic strategies.


Subject(s)
Immunotherapy , Melanoma, Experimental/therapy , Peptide Fragments/pharmacology , Transforming Growth Factor beta/antagonists & inhibitors , Adjuvants, Immunologic/therapeutic use , Animals , CD40 Antigens/immunology , Dendritic Cells/immunology , Female , Flow Cytometry , Homeodomain Proteins/physiology , Humans , Killer Cells, Natural/immunology , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Poly I-C/immunology , Poly I-C/therapeutic use , Receptors, Transforming Growth Factor beta , Survival Rate , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Regulatory/immunology , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...