Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Methods Mol Biol ; 1880: 455-477, 2019.
Article in English | MEDLINE | ID: mdl-30610715

ABSTRACT

Macroautophagy is a ubiquitous degradative pathway involved in innate and adaptive immunity. Its molecular machinery has been described to deliver intracellular and extracellular antigens to MHC class II loading compartment by regulating autophagosome and phagosome maturation. We recently found that the respective Atg proteins can contribute to MHC class I-restricted antigen presentation to CD8+ T cells by regulating MHC class I surface levels in mouse dendritic cell. Indeed, we determined that MHC class I molecules are stabilized on the cell surface of murine antigen presenting cells deficient for core components of the macroautophagy machinery such as Atg5 and Atg7. This stabilization seems to result from defective internalization of MHC class I molecules dependent on adaptor protein kinase 1 (AAK1), involved in clathrin-mediated endocytosis. Moreover, macroautophagy-dependent stabilization of MHC class I molecules leads to enhanced CD8+ T cell priming during influenza A virus infection in vivo, resulting in decreased pathology. In this chapter, we describe four experiments to monitor, characterize, and quantify the effect of macroautophagy deficiency on MHC class I molecule trafficking and the subsequent CD8+ T cell priming. First, we will show how to monitor MHC class I internalization in lung CD11c+ cells from mice lacking key components of the macroautophagy machinery. Then, we will propose a method to characterize the interaction between either MHC class I or Atg8/LC3 with AAK1. Finally, we will describe how to evaluate the influenza A-specific CD8+ T cell response in mice conditionally depleted for Atg5 in their DC compartment. This set of experiments allows to characterize MHC class I internalization with the help of the molecular machinery of macroautophagy.


Subject(s)
Autophagy-Related Proteins/metabolism , Autophagy/immunology , Biological Assay/methods , Histocompatibility Antigens Class I/metabolism , Animals , Antigen Presentation/immunology , Autophagosomes/metabolism , Autophagy-Related Proteins/genetics , Autophagy-Related Proteins/immunology , Biological Assay/instrumentation , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/microbiology , Cell Separation/instrumentation , Cell Separation/methods , Dendritic Cells/immunology , Dendritic Cells/metabolism , Dendritic Cells/microbiology , Endocytosis/immunology , Histocompatibility Antigens Class I/immunology , Influenza A virus/immunology , Lung/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Fluorescence/instrumentation , Microscopy, Fluorescence/methods , Microtubule-Associated Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism
2.
Curr Protoc Immunol ; 123(1): e60, 2018 11.
Article in English | MEDLINE | ID: mdl-30253073

ABSTRACT

LC3-associated phagocytosis (LAP) is an unconventional form of autophagy that relies on parts of the canonical autophagy machinery for its function. LAP is triggered upon receptor-mediated phagocytosis and is characterized by the formation of a single-membrane vesicle decorated with the autophagy protein LC3. In professional phagocytic cells, such as macrophages, the role of LAP in immune processes has been characterized, although how LAP functions at the molecular level remains poorly defined. It is important to point out that as for all autophagic pathways, the study of LAP is still challenging for the scientific community because it is a dynamic and complex process, requiring interactions among several proteins. Here, we describe the most common methods used to monitor and quantify the formation of LC3-coated single-membrane endosomes, or so-called LAPosomes, and to validate the involvement of LAP in immunological processes of human macrophages. © 2018 by John Wiley & Sons, Inc.


Subject(s)
Antigen Presentation , Endosomes/immunology , Macrophages/immunology , Microtubule-Associated Proteins/immunology , Phagocytosis , Animals , Humans
3.
Curr Opin Immunol ; 52: 68-73, 2018 06.
Article in English | MEDLINE | ID: mdl-29719275

ABSTRACT

The macroautophagy machinery supports membrane remodeling and fusion events that lead to the engulfment of cytoplasmic constituents in autophagosomes and their degradation in lysosomes. The capacity of this machinery to regulate membrane adaptors and influence vesicle fusion with lysosomes seems to be used not only for autophagosomes, but also for endosomes. We summarize recent evidence that two aspects of endocytosis are regulated by parts of the macroautophagy machinery. These are recruitment of adaptors for the internalization of surface receptors and the fusion of phagosomes with lysosomes. Antigen processing for MHC presentation is affected by these alternative functions of the macroautophagy machinery. Primarily extracellular antigen presentation by MHC class II molecules after phagocytosis benefits from this regulation of phagosome maturation. Furthermore, MHC class I molecules are more efficiently internalized in the presence of the core macroautophagy machinery. The identification of these alternative functions of macroautophagy proteins not only complicates the interpretation of their deficiencies in biological processes, but could also be harnessed for the regulation of antigen presentation to T cells.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Autophagy-Related Proteins/genetics , Endocytosis/immunology , Histocompatibility Antigens/immunology , Animals , Antigens/metabolism , Autophagy/genetics , Autophagy/immunology , Autophagy-Related Proteins/metabolism , Histocompatibility Antigens/metabolism , Humans , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Phagocytes/immunology , Phagocytes/metabolism , Phagocytosis/genetics , Phagocytosis/immunology , Phagosomes/immunology , Phagosomes/metabolism
4.
Autophagy ; 13(6): 1025-1036, 2017 Jun 03.
Article in English | MEDLINE | ID: mdl-28296542

ABSTRACT

Invariant natural killer T (iNKT) cells are innate T cells with powerful immune regulatory functions that recognize glycolipid antigens presented by the CD1D protein. While iNKT cell-activating glycolipids are currently being explored for their efficacy to improve immunotherapy against infectious diseases and cancer, little is known about the mechanisms that control CD1D antigen presentation and iNKT cell activation in vivo. CD1D molecules survey endocytic pathways to bind lipid antigens in MHC class II-containing compartments (MIICs) before recycling to the plasma membrane. Autophagosomes intersect with MIICs and autophagy-related proteins are known to support antigen loading for increased CD4+ T cell immunity. Here, we report that mice with dendritic cell (DC)-specific deletion of the essential autophagy gene Atg5 showed better CD1D1-restricted glycolipid presentation in vivo. These effects led to enhanced iNKT cell cytokine production upon antigen recognition and lower bacterial loads during Sphingomonas paucimobilis infection. Enhanced iNKT cell activation was independent of receptor-mediated glycolipid uptake or costimulatory signals. Instead, loss of Atg5 in DCs impaired clathrin-dependent internalization of CD1D1 molecules via the adaptor protein complex 2 (AP2) and, thus, increased surface expression of stimulatory CD1D1-glycolipid complexes. These findings indicate that the autophagic machinery assists in the recruitment of AP2 to CD1D1 molecules resulting in attenuated iNKT cell activation, in contrast to the supporting role of macroautophagy in CD4+ T cell stimulation.


Subject(s)
Antigens, CD1d/metabolism , Autophagy , Endocytosis , Natural Killer T-Cells/cytology , Natural Killer T-Cells/metabolism , Adaptor Protein Complex 2/metabolism , Animals , Antigens/metabolism , Autophagy-Related Protein 5/metabolism , Bacterial Infections/metabolism , Bacterial Infections/pathology , Cell Membrane/metabolism , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/metabolism , Endosomes/metabolism , Glycolipids/metabolism , Immunization , Lipids/immunology , Mice, Inbred C57BL , Mice, Knockout
5.
Autophagy ; 12(9): 1681-2, 2016 09.
Article in English | MEDLINE | ID: mdl-27439741

ABSTRACT

We have recently shown that the LC3/Atg8 lipidation machinery of macroautophagy is involved in the internalization of MHC class I molecules. Decreased internalization in the absence of ATG5 or ATG7 leads to MHC class I surface stabilization on dendritic cells and macrophages, resulting in elevated CD8(+) T cell responses during viral infections and improved immune control. Here, we discuss how the autophagic machinery supports MHC class II restricted antigen presentation, while compromising MHC class I presentation via internalization and degradation.


Subject(s)
Antigen Presentation/immunology , Autophagy-Related Protein 5/metabolism , Autophagy-Related Protein 7/metabolism , Autophagy , Histocompatibility Antigens Class II/metabolism , Animals , CD8-Positive T-Lymphocytes/cytology , Cell Membrane/metabolism , Cell-Free System , Dendritic Cells/metabolism , Humans , Macrophages/metabolism , Phagocytosis , T-Lymphocytes/immunology
6.
Cell Rep ; 15(5): 1076-1087, 2016 05 03.
Article in English | MEDLINE | ID: mdl-27117419

ABSTRACT

The macroautophagy machinery has been implicated in MHC class II restricted antigen presentation. Here, we report that this machinery assists in the internalization of MHC class I molecules. In the absence of the autophagy factors Atg5 and Atg7, MHC class I surface levels are elevated due to decreased endocytosis and degradation. Internalization of MHC class I molecules occurs less efficiently if AAK1 cannot be recruited via Atg8/LC3B. In the absence of Atg-dependent MHC class I internalization, dendritic cells stimulate CD8(+) T cell responses more efficiently in vitro and in vivo. During viral infections, lack of Atg5 results in enhanced influenza- and LCMV-specific CD8(+) T cell responses in vivo. Elevated influenza-specific CD8(+) T cell responses are associated with better immune control of this infection. Thus, the macroautophagy machinery orchestrates T cell immunity by supporting MHC class II but compromises MHC class I restricted antigen presentation.


Subject(s)
Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 7/genetics , Autophagy/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Histocompatibility Antigens Class I/immunology , Influenza A Virus, H1N1 Subtype/immunology , Lymphocytic choriomeningitis virus/immunology , Animals , Antigen Presentation/immunology , Cells, Cultured , Endocytosis/immunology , Histocompatibility Antigens Class II/immunology , Humans , Lymphocyte Activation/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout
7.
Biomaterials ; 68: 89-99, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26276694

ABSTRACT

Selective tumor targeting is expected to enhance drug delivery and to decrease toxicity, resulting in an improved therapeutic index. We have recently identified the HSYWLRS peptide sequence as a specific ligand for aggressive neuroblastoma, a childhood tumor mostly refractory to current therapies. Here we validated the specific binding of HSYWLRS to neuroblastoma cell suspensions obtained either from cell lines, animal models, or Schwannian-stroma poor, stage IV neuroblastoma patients. Binding of the biotinylated peptide and of HSYWLRS-functionalized fluorescent quantum dots or liposomal nanoparticles was dose-dependent and inhibited by an excess of free peptide. In animal models obtained by the orthotopic implant of either MYCN-amplified or MYCN single copy human neuroblastoma cell lines, treatment with HSYWLRS-targeted, doxorubicin-loaded Stealth Liposomes increased tumor vascular permeability and perfusion, enhancing tumor penetration of the drug. This formulation proved to exert a potent antitumor efficacy, as evaluated by bioluminescence imaging and micro-PET, leading to (i) delay of tumor growth paralleled by decreased tumor glucose consumption, and (ii) abrogation of metastatic spreading, accompanied by absence of systemic toxicity and significant increase in the animal life span. Our findings are functional to the design of targeted nanocarriers with potentiated therapeutic efficacy towards the clinical translation.


Subject(s)
Doxorubicin/administration & dosage , Nanocapsules/administration & dosage , Neoplasm Metastasis/prevention & control , Neuroblastoma/chemistry , Neuroblastoma/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Diffusion , Doxorubicin/chemistry , Drug Synergism , Female , Mice , Mice, Nude , Nanocapsules/chemistry , Neoplasm Invasiveness , Neoplasm Metastasis/pathology , Neuroblastoma/pathology
8.
J Control Release ; 211: 44-52, 2015 Aug 10.
Article in English | MEDLINE | ID: mdl-26031842

ABSTRACT

Neuroblastoma is a childhood cancer with poor long-term prognosis in advanced stages. A major aim in neuroblastoma therapy is to develop targeted drug delivery systems to ameliorate drug therapeutic index and efficacy. In this study, a novel bortezomib (BTZ) liposomal formulation was set-up and characterized. Since BTZ is freely permeable across the lipidic bilayer, an amino-lactose (LM) was synthesized as complexing agent to entrap BTZ inside the internal aqueous compartment of stealth liposomes. High encapsulation efficiency was achieved by a loading method based on the formation of boronic esters between the boronic acid moiety of BTZ and the hydroxyl groups of LM. Next, NGR peptides were linked to the liposome surface as a targeting-ligand for the tumor endothelial cell marker, aminopeptidase N. Liposomes were characterized for size, Z-potential, polydispersity index, drug content, and release. Lyophilization in the presence of cryoprotectants (trehalose, sucrose) was also examined in terms of particle size changes and drug leakage. BTZ was successfully loaded into non-targeted (SL[LM-BTZ]) and targeted (NGR-SL[LM-BTZ]) liposomes with an entrapment efficiency of about 68% and 57%, respectively. These nanoparticles were suitable for intravenous administration, presenting an average diameter of 170nm and narrow polydispersity. Therefore, orthotopic NB-bearing mice were treated with 1.0 or 1.5mg/kg of BTZ, either in free form or encapsulated into liposomes. BTZ loaded liposomes showed a significant reduction of drug systemic adverse effects with respect to free drug, even at the highest dose tested. Moreover, mice treated with 1.5mg/kg of NGR-SL[LM-BTZ] lived statistically longer than untreated mice (P=0.0018) and SL[LM-BTZ]-treated mice (P=0.0256). Our results demonstrate that the novel vascular targeted BTZ formulation is endowed with high therapeutic index and low toxicity, providing a new tool for future applications in neuroblastoma clinical studies.


Subject(s)
Antineoplastic Agents/administration & dosage , Bortezomib/administration & dosage , Drug Delivery Systems/methods , Neovascularization, Pathologic/drug therapy , Neuroblastoma/drug therapy , Animals , Cell Line, Tumor , Chemistry, Pharmaceutical , Dose-Response Relationship, Drug , Female , Humans , Liposomes , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Neuroblastoma/pathology , Xenograft Model Antitumor Assays/methods
9.
Front Oncol ; 3: 190, 2013.
Article in English | MEDLINE | ID: mdl-23936762

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor in children, accounting for about 8% of childhood cancers. Despite aggressive treatment, patients suffering from high-risk NB have very poor 5-year overall survival rate, due to relapsed and/or treatment-resistant tumors. A further increase in therapeutic dose intensity is not feasible, because it will lead to prohibitive short-term and long-term toxicities. New approaches with targeted therapies may improve efficacy and decrease toxicity. The use of drug delivery systems allows site specific delivery of higher payload of active agents associated with lower systemic toxicity compared to the use of conventional ("free") drugs. The possibility of imparting selectivity to the carriers to the cancer foci through the use of a targeting moiety (e.g., a peptide or an antibody) further enhances drug efficacy and safety. We have recently developed two strategies for increasing local concentration of anti-cancer agents, such as CpG-containing oligonucleotides, small interfering RNAs, and chemotherapeutics in NB. For doing that, we have used the monoclonal antibody anti-disialoganglioside (GD2), able to specifically recognize the NB tumor and the peptides containing NGR and CPRECES motifs, that selectively bind to the aminopeptidase N-expressing endothelial and the aminopeptidase A-expressing perivascular tumor cells, respectively. The review will focus on the use of tumor- and tumor vasculature-targeted nanocarriers to improve tumor targeting, uptake, and penetration of drugs in preclinical models of human NB.

10.
J Control Release ; 170(3): 445-51, 2013 Sep 28.
Article in English | MEDLINE | ID: mdl-23792118

ABSTRACT

Neuroblastoma is an embryonal tumor originating from the simpatico-adrenal lineage of the neural crest. It approximately accounts for about 15% of all pediatric oncology deaths. Despite advances in multimodal therapy, metastatic neuroblastoma tumors at diagnosis remain a clinical challenge. Retinoids are a class of compounds known to induce both terminal differentiation and apoptosis/necrosis of neuroblastoma cells. Among them, fenretinide (HPR) has been considered one of the most promising anti-tumor agent but it is partially efficacious due to both poor aqueous solubility and rapid metabolism. Here, we have developed a novel HPR formulation, by which the drug was encapsulated into sterically stabilized nanoliposomes (NL[HPR]) according to the Reverse Phase Evaporation method. This procedure led to a higher structural integrity of liposomes in organic fluids for a longer period of time, in comparison with our previous liposomal formulation developed by the film method. Moreover, NL[HPR] were further coupled with NGR peptides for targeting the tumor endothelial cell marker, aminopeptidase N (NGR-NL[HPR]). Orthotopically xenografted neuroblastoma-bearing mice treated with NGR-NL[HPR] lived statistically longer than mice untreated or treated with free HPR (NGR-NL[HPR] vs both control and HPR: P<0.0001). Also, NL[HPR] resulted in a statistically improved survival (NL[HPR] vs both control and HPR: P<0.001) but to a less extent if compared with that obtained with NGR-NL[HPR] (NGR-NL[HPR] vs NL[HPR]: P<0.01). Staining of tumor sections with antibodies specific for neuroblastoma and for either pericytes or endothelial cells evidenced that HPR reduced neuroblastoma growth through both anti-tumor and anti-angiogenic effects, mainly when delivered by NGR-NL[HPR]. Indeed, in this group of mice a marked reduction of tumor progression, of intra-tumoral vessel counts and VEGF expression, together with a marked down-modulation of matrix metalloproteinases MMP2 and MMP9, was observed. In conclusion, the use of this novel targeted delivery system for the apoptotic and antiangiogenic drug, fenretinide, could be considered as an adjuvant tool in the future treatment of neuroblastoma patients.


Subject(s)
Antineoplastic Agents/administration & dosage , Fenretinide/administration & dosage , Neovascularization, Pathologic/drug therapy , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/chemistry , Cell Line, Tumor , Female , Fenretinide/chemistry , Humans , Liposomes , Mice , Mice, Nude , Neovascularization, Pathologic/pathology , Neuroblastoma/pathology
11.
J Control Release ; 170(2): 233-41, 2013 Sep 10.
Article in English | MEDLINE | ID: mdl-23714122

ABSTRACT

Molecular targeting of drug delivery nanocarriers is expected to improve their therapeutic index while decreasing their toxicity. Here we report the identification and characterization of novel peptide ligands specific for cells present in high-risk neuroblastoma (NB), a childhood tumor mostly refractory to current therapies. To isolate such targeting moieties, we performed combined in vitro/ex-vivo phage display screenings on NB cell lines and on tumors derived from orthotopic mouse models of human NB. By designing proper subtractive protocols, we identified phage clones specific either for the primary tumor, its metastases, or for their respective stromal components. Globally, we isolated 121 phage-displayed NB-binding peptides: 26 bound the primary tumor, 15 the metastatic mass, 57 and 23 their respective microenvironments. Of these, five phage clones were further validated for their specific binding ex-vivo to biopsies from stage IV NB patients and to NB tumors derived from mice. All five clones also targeted tumor cells and vasculature in vivo when injected into NB-bearing mice. Coupling of the corresponding targeting peptides with doxorubicin-loaded liposomes led to a significant inhibition in tumor volume and enhanced survival in preclinical NB models, thereby paving the way to their clinical development.


Subject(s)
Doxorubicin/administration & dosage , Nanoparticles/administration & dosage , Neuroblastoma/drug therapy , Peptides/administration & dosage , Animals , Cell Line, Tumor , Cell Surface Display Techniques , Cell Survival/drug effects , Cells, Cultured , Doxorubicin/chemistry , Female , Human Umbilical Vein Endothelial Cells , Humans , Liposomes , Mice , Mice, Nude , Nanoparticles/chemistry , Neuroblastoma/pathology , Peptides/chemistry , Peptides/pharmacokinetics , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
12.
Int J Dev Biol ; 55(4-5): 547-55, 2011.
Article in English | MEDLINE | ID: mdl-21858775

ABSTRACT

The orthotopic model reproduces aspects of the tumour microenvironment and emulates a number of important biological features of cancer progression, angiogenesis, metastasis and resistance. Due to its parallels with human cancer, the model can be used to evaluate therapeutic responses to various therapies. This review outlines the importance of using the orthotopic implantation of tumour cells in mice models for evaluating the effectiveness of antivascular therapies.


Subject(s)
Angiogenesis Inhibitors/therapeutic use , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/drug therapy , Angiogenesis Inhibitors/administration & dosage , Animals , Antineoplastic Agents/administration & dosage , Disease Models, Animal , Humans , Mice , Neoplasm Transplantation/methods , Neovascularization, Pathologic/therapy , Tumor Microenvironment , Xenograft Model Antitumor Assays
13.
Mol Ther ; 19(12): 2201-12, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21829174

ABSTRACT

The anaplastic lymphoma kinase (ALK) is a tyrosine kinase receptor that is involved in the pathogenesis of different types of human cancers, including neuroblastoma (NB). In NB, ALK overexpression, or point mutations, are associated with poor prognosis and advanced stage disease. Inhibition of ALK kinase activity by small-molecule inhibitors in lung cancers carrying ALK translocations has shown therapeutic potential. However, secondary mutations may occur that, generate tumor resistance to ALK inhibitors. To overcome resistance to ALK inhibitors in NB, we adopted an alternative RNA interference (RNAi)-based therapeutic strategy that is able to knockdown ALK, regardless of its genetic status [mutated, amplified, wild-type (WT)]. NB cell lines, transduced by lentiviral short hairpin RNA (shRNA), showed reduced proliferation and increased apoptosis when ALK was knocked down. In mice, a nanodelivery system for ALK-specific small interfering RNA (siRNA), based on the conjugation of antibodies directed against the NB-selective marker GD(2) to liposomes, showed strong ALK knockdown in vivo in NB cells, which resulted in cell growth arrest, apoptosis, and prolonged survival. ALK knockdown was associated with marked reductions in vascular endothelial growth factor (VEGF) secretion, blood vessel density, and matrix metalloproteinases (MMPs) expression in vivo, suggesting a role for ALK in NB-induced neoangiogenesis and tumor invasion, confirming this gene as a fundamental oncogene in NB.


Subject(s)
Apoptosis , Mutation/genetics , Neovascularization, Pathologic/prevention & control , Neuroblastoma/blood supply , Neuroblastoma/therapy , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/antagonists & inhibitors , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Female , Gangliosides/immunology , Gangliosides/metabolism , HeLa Cells , Humans , Immunoenzyme Techniques , Liposomes , Matrix Metalloproteinases/genetics , Matrix Metalloproteinases/metabolism , Mice , Mice, Nude , Mice, SCID , Neuroblastoma/mortality , Phosphorylation , RNA Interference , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Signal Transduction , Survival Rate
14.
Mol Ther ; 19(6): 1131-40, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21487394

ABSTRACT

RNA interference molecules have some advantages as cancer therapeutics, including a proved efficacy on both wild-type (WT) and mutated transcripts and an extremely high sequence-specificity. The most significant hurdle to be overcome if exogenous small interfering RNAs (siRNA) is to be used therapeutically is the specific, effective, nontoxic delivery of siRNA to its intracellular site of action. At present, human applications are confined almost exclusively to targets within the liver, where the delivery systems naturally accumulate, and extra-hepatic targets remain a challenge. Anaplastic lymphoma kinase (ALK) is a receptor tyrosine kinase that has recently been shown to contribute to the cell growth and progression of human neuroblastoma (NB). We investigated its potential as a therapeutic target in NB by generating anti-GD2-targeted nanoparticles that carry ALK-directed siRNA, which are specifically and efficiently delivered to GD2-expressing NB cells. Relative to free ALK-siRNA, anti-GD2-targeted liposomal formulations of ALK-siRNA had low plasma clearance, increased siRNA stability, and improved binding, uptake, silencing and induction of cell death, and specificity for NB cells. In NB xenografts, intravenous (i.v.) injection of the targeted ALK-siRNA liposomes showed gene-specific antitumor activity with no side effects. ALK-selective siRNA entrapped in anti-GD2-targeted nanoparticles is a promising new modality for NB treatment.


Subject(s)
Neuroblastoma/enzymology , Neuroblastoma/therapy , RNA, Small Interfering/physiology , Receptor Protein-Tyrosine Kinases/metabolism , Anaplastic Lymphoma Kinase , Animals , Blotting, Western , Cell Line , Gene Silencing/physiology , Humans , Mice , Mice, Nude , Nanoparticles/chemistry , Neuroblastoma/genetics , RNA, Small Interfering/genetics , Receptor Protein-Tyrosine Kinases/genetics , Reverse Transcriptase Polymerase Chain Reaction , Transfection , Xenograft Model Antitumor Assays
15.
Cancer Res ; 70(23): 9816-26, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20935225

ABSTRACT

The Toll-like receptor 9 (TLR9) evolved to cope with pathogens, but it is expressed in a variety of tumors for reasons that are unclear. In this study, we report that neuroblastoma (NB) cells express functional TLR9. Liposome-complexed CpG oligonucleotides inhibited the proliferation of TLR9-expressing NB cells and induced caspase-dependent apoptotic cell death. Inhibitory oligonucleotides (iODNs) abrogated these effects. RNA interference reduced TLR9 expression but not to the level where functional responses to CpG were abolished. Compared with free CpG, liposomal formulations of NB-targeted CpG (TL-CpG) significantly prolonged the survival of mice bearing NB tumor xenografts. While CpG alone lacked antitumor efficacy in NOD/SCID/IL2rg(-/-) mice, TL-CpG retained significant efficacy related to direct effects on tumor cells. TLR9 expression in primary human NB specimens was found to correlate inversely with disease stage. Our findings establish functional expression of TLR9 in NB and suggest that TLR9 may represent a novel theranostic target in this disease.


Subject(s)
Apoptosis/drug effects , Cell Proliferation/drug effects , Neuroblastoma/therapy , Oligodeoxyribonucleotides/pharmacology , RNA Interference , Toll-Like Receptor 9/antagonists & inhibitors , Animals , Blotting, Western , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Humans , Infant , Interleukin Receptor Common gamma Subunit/deficiency , Interleukin Receptor Common gamma Subunit/genetics , Liposomes/chemistry , Mice , Mice, Inbred NOD , Mice, Knockout , Mice, Nude , Mice, SCID , Neoplasm Staging , Neuroblastoma/genetics , Neuroblastoma/pathology , Oligodeoxyribonucleotides/genetics , Oligodeoxyribonucleotides, Antisense/chemistry , Oligodeoxyribonucleotides, Antisense/genetics , Oligodeoxyribonucleotides, Antisense/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Survival Analysis , Toll-Like Receptor 9/genetics , Toll-Like Receptor 9/metabolism , Xenograft Model Antitumor Assays
16.
Clin Cancer Res ; 16(19): 4809-21, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20702613

ABSTRACT

PURPOSE: Treatment of neuroblastoma is successful in less than half of patients with high-risk disease. The antitumor activity of a water soluble pegylated SN38 drug conjugate, EZN-2208, was compared with CPT-11 (a prodrug for SN38) in preclinical models of human neuroblastoma. EXPERIMENTAL DESIGN: The in vitro cytotoxicity of EZN-2208 was tested by counting trypan blue dye- and Annexin V-positive cells, whereas its therapeutic efficacy was evaluated, in terms of survival, and antitumor and antiangiogenic activities, in s.c. luciferase-transfected, pseudometastatic, and orthotopic neuroblastoma animal models. RESULTS: EZN-2208 was about 100-fold more potent than CPT-11 in vitro, by inducing apoptosis/necrosis and p53 expression and by reducing hypoxia-inducible factor (HIF)-1α/HIF-2α expression. EZN-2208 gave superior antitumor effects compared with CPT-11 in neuroblastoma xenografts. EZN-2208 treatment always resulted in lack of tumor detection at the end of trials whereas only small therapeutic effects were observed with CPT-11, as assessed by luciferase assay or tumor size, or even by staining histologic sections of tumors with antibodies recognizing neuroblastoma cells and cell proliferation. In a neuroblastoma model resistant to doxorubicin, cisplatin, vincristine, fenretinide, and topotecan, EZN-2208 induced 100% curability. It also blocked tumor relapse after topotecan-vincristine-doxorubicin combined treatment. Mechanistic experiments showed statistically significantly enhanced terminal deoxynucleotidyl transferase-mediated dUTP nick end labeling and Histone H2ax staining as well as decreased vascular endothelial growth factor, CD31, matrix metalloproteinase (MMP)-2, and MMP-9 expression in tumors removed from EZN-2208-treated mice and radiating vessels invading the tumor implanted onto the chorioallantoic membranes. CONCLUSIONS: EZN-2208 should be considered a most promising novel antineuroblastoma agent. An ongoing phase I study in pediatric patients should identify the optimal dose for a phase II study.


Subject(s)
Antineoplastic Agents/pharmacology , Camptothecin/analogs & derivatives , Disease Models, Animal , Neoplasms, Experimental/drug therapy , Neuroblastoma/drug therapy , Polyethylene Glycols/pharmacology , Polyethylene Glycols/therapeutic use , Topoisomerase I Inhibitors/pharmacology , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Camptothecin/pharmacology , Camptothecin/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Topoisomerases, Type I/metabolism , Drug Screening Assays, Antitumor , Female , Humans , Irinotecan , Mice , Mice, Nude , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/drug therapy , Neuroblastoma/pathology , Survival Rate , Topoisomerase I Inhibitors/therapeutic use
17.
J Control Release ; 145(1): 66-73, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20346382

ABSTRACT

The therapeutic index of anti-cancer drugs is increased when encapsulating them in tumor-targeted liposomes. Liposome-entrapped doxorubicin (DXR), targeting the tumor vasculature marker, aminopeptidase N (APN), displayed enhanced anti-tumor effects and prolonged survival in human neuroblastoma (NB)-bearing mice. Here we exploited a peptide ligand of aminopeptidase A (APA), discovered by phage display technology for delivery of liposomal DXR to perivascular tumor cells. Immunohistochemistry, performed in NB-bearing mice, showed APA expression in the vascular wall of NB primary and metastatic lesions. APA-targeted peptides displayed specific binding to APA-transfected cells in vitro, and also accumulation in the tumor of NB-bearing mice. Consequently, novel, APA-targeted, DXR-liposomes were developed and in vivo proof-of-principle was established, alone and in combination with APN-targeted DXR-loaded liposomes, in NB-bearing mice. Mice receiving APA-targeted liposomal DXR exhibited an increased life span in comparison to control mice, but to a lesser extent relative to that in mice treated with APN-targeted formulation, moreover the greatest increase in TUNEL-positive tumor cells was observed in animals treated with APN-targeted formulations. Mice treated with a combination of APA- and APN-targeted, liposomal DXR had a significant increase in life span compared to each treatment administered separately. There was a significant increase in the level of apoptosis in the tumors of mice on the combination therapy, and a pronounced destruction of the tumor vasculature with nearly total ablation of endothelial cells and pericytes. The availability of novel ligands binding to additional tumor vasculature-associated antigens will allow the design of sophisticated combinations of ligand-targeted liposomal anti-cancer drugs.


Subject(s)
Antibiotics, Antineoplastic/therapeutic use , Doxorubicin/therapeutic use , Endothelial Cells/drug effects , Neuroblastoma/drug therapy , Animals , Antibiotics, Antineoplastic/administration & dosage , Antibiotics, Antineoplastic/pharmacokinetics , CD13 Antigens/biosynthesis , CD13 Antigens/chemistry , Doxorubicin/administration & dosage , Doxorubicin/pharmacokinetics , Drug Compounding , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Glutamyl Aminopeptidase/biosynthesis , Glutamyl Aminopeptidase/chemistry , Humans , Immunohistochemistry , Ligands , Liposomes , Mice , Mice, Nude , Neuroblastoma/blood supply , Neuroblastoma/metabolism , Neuroblastoma/pathology , Peptides/chemistry , Peptides/metabolism , Protein Binding , Xenograft Model Antitumor Assays
18.
Methods Enzymol ; 465: 225-49, 2009.
Article in English | MEDLINE | ID: mdl-19913170

ABSTRACT

Neuroblastoma (NB) is the most common extracranial solid tumor in childhood and the most frequently diagnosed neoplasm during infancy. Despite of aggressive treatment strategies, the 5-year survival rate for metastatic disease is still less than 60% and, consequently, novel therapeutic approaches are needed. For increasing the therapeutic index of anticancer drugs, while reducing side effects, one of the most promising strategies in modern chemotherapy is based on the development of innovative drug delivery systems, such as liposomes. "Anticancer drug"-loaded liposomes have demonstrated enhanced ability to target to the affected area, as well as increased antitumor efficacy compared to conventional drugs. Liposomes tend to extravasate preferentially and to accumulate into tumor interstitial fluids, due to the defective structure of the new angiogenic vessels within the tumor masses. This inherent tumor selectivity can be increased further by coupling tumor-specific antibodies or other targeting moieties to the surface of the lipid envelope. Here, we describe the methodology used in these studies, as well as the antitumor results obtained by the use of several "anticancer drugs," encapsulated into antibody- and peptide-targeted liposomal formulations, against NB.


Subject(s)
Antineoplastic Agents/therapeutic use , Doxorubicin/therapeutic use , Fenretinide/therapeutic use , Liposomes , Neuroblastoma/drug therapy , Animals , Antineoplastic Agents/administration & dosage , Doxorubicin/administration & dosage , Drug Delivery Systems , Fenretinide/administration & dosage , Gangliosides/chemistry , Gold , Mice , Microscopy, Electron, Transmission , Neoplasm Transplantation , Neovascularization, Pathologic/drug therapy , Neuroblastoma/blood supply
19.
J Control Release ; 138(2): 122-7, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19427884

ABSTRACT

High-risk Neuroblastoma (NB) has still a poor prognosis. Liposomes targeted to NB cells and encapsulating antisense CpG-containing oligonucleotides (TL-asCpG) had increased anti-tumour efficacy in NB xenografts compared to free asCpG. Interleukin 10 (IL-10) suppresses antigen presenting cell activation contributing to tumour-mediated immune suppression. In principle, combination of TL-asCpG and antibodies against IL-10 receptor (aIL-10R) could prolong immune system activation, leading to better therapeutic results. Mice treated with TL-asCpG 4 h after human NB cell inoculation survived significantly longer than controls. An increased life span was achieved also in mice receiving TL-asCpG 24 and 72 h after NB cell challenge. The addition of aIL-10R to TL-asCpG in the 4-h protocol significantly increased the percentage of long term survivors compared to TL-asCpG only. Surviving mice treated with the combined strategy were completely cured. In contrast, long term surviving mice treated only with TL-asCpG presented lymph node infiltration with NB cells. TL-asCpG plus aIL-10R treatment was significantly superior to TL-asCpG alone also for the 24-h protocol. Ex vivo experiments demonstrated that the combined therapy evoked a stronger and more prolonged immune system activation compared to monotherapy. These results support the feasibility of a clinical trial with TL-asCpG and aIL-10R in advanced NB patients.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Immunity, Innate/drug effects , Interleukin-10/antagonists & inhibitors , Neuroblastoma/prevention & control , Oligonucleotides, Antisense/therapeutic use , Receptors, Interleukin-10/antagonists & inhibitors , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Cell Line, Tumor , Cell Proliferation/drug effects , CpG Islands/immunology , Female , Gangliosides/immunology , Humans , Interleukin-10/immunology , Liposomes , Mice , Mice, Nude , Neoplasm Transplantation , Neuroblastoma/immunology , Neuroblastoma/pathology , Oligonucleotides, Antisense/administration & dosage , Oligonucleotides, Antisense/immunology , Proto-Oncogene Proteins c-myb/immunology , Receptors, Interleukin-10/immunology , Xenograft Model Antitumor Assays
20.
Clin Cancer Res ; 15(4): 1199-209, 2009 Feb 15.
Article in English | MEDLINE | ID: mdl-19228726

ABSTRACT

PURPOSE: The proteasome inhibitor bortezomib inhibited cell growth and angiogenesis in neuroblastoma. Bortezomib has been shown to induce synergistic activity when combined with other antineoplastic agents. Here we have investigated the antitumor activity of bortezomib in combination with fenretinide, a synthetic retinoid, against neuroblastoma cells. EXPERIMENTAL DESIGN: Different neuroblastoma cell lines were tested for sensitivity to bortezomib and fenretinide, given alone or in different dose-dependent and time-dependent combination schedules. Cell proliferation, cell viability, and apoptosis were evaluated by measuring 3H-thymidine incorporation, trypan blue staining, DNA fragmentation, and western blot analysis. Angiogenesis was assessed by the chick embryo chorioallantoic membrane assay. An orthotopic neuroblastoma mouse model was used to examine in vivo sensitivity. RESULTS: Each compound alone was able to induce a dose-dependent inhibition of cell proliferation, with a significant enhanced antiproliferative effect for the drugs used in combination. This inhibition was characterized by marked G2-M and G1 cell cycle arrest with nearly complete depletion of S phase. Bortezomib and fenretinide in association triggered an increased apoptosis through activation of specific genes of the endoplasmic reticulum stress compared with either drug tested alone. Tumor-bearing mice treated with bortezomib plus fenretinide lived statistically significantly longer than mice treated with each drug alone. Histologic evaluation and chorioallantoic membrane analysis of primary tumors showed that the combined therapeutic activity of bortezomib and fenretinide rested upon antitumor and antiangiogenic mechanisms. CONCLUSIONS: These findings provide the rationale for the development of a new therapeutic strategy for neuroblastoma based on this pharmacologic combination.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Boronic Acids/administration & dosage , Endoplasmic Reticulum/drug effects , Fenretinide/administration & dosage , Neuroblastoma/drug therapy , Pyrazines/administration & dosage , Animals , Apoptosis/drug effects , Bortezomib , Cell Line, Tumor , Cell Proliferation/drug effects , Chick Embryo , Endoplasmic Reticulum/metabolism , Humans , Mice , Neuroblastoma/mortality , Neuroblastoma/pathology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...