Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 78
1.
Int J Biol Macromol ; 183: 340-345, 2021 Jul 31.
Article En | MEDLINE | ID: mdl-33932411

Yellowhead catfish (Tachysurus fulvidraco) is an important aquaculture fish species in China with a high market value. Infectious diseases pose serious threats in farmed fish species, and although vaccines can prevent certain infections, they rely on potent adjuvants. In this study, we analyzed the transcriptomic profiles of spleens from poly (I:C)-treated T. fulvidraco. We obtained 46,362,922 reads corresponding to 490,926 transcripts and 318,059 genes. Gene annotation using different databases and subsequent differential gene expression analyses led to the identification of 5587 differentially expressed genes (DEGs), of which 2473 were up-regulated and 3114 were down-regulated in poly (I:C)-treated fish. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses of DEGs revealed the significant dysregulation of immune- and cancer-related genes in the spleens of poly (I:C)-treated fish. Notably, several components of JAK-STAT, MAPK, and p53 signaling pathways were significantly dysregulated in response to poly (I:C) treatment. Quantitative real-time PCR (qRT-PCR) analysis of 11 randomly selected immune response genes confirmed the reliability of our findings. In conclusion, our findings provide novel insight into the immune responses of T. fulvidraco and suggest that poly (I:C) may represent a promising adjuvant of fish vaccines.


Poly I-C/chemistry , Animals , Catfishes , Gene Expression Profiling , Transcriptome/genetics
2.
ACS Appl Mater Interfaces ; 12(47): 52285-52297, 2020 Nov 25.
Article En | MEDLINE | ID: mdl-33179910

In this study, biodegradable cationic polycarbonate and polylactide block copolymers were synthesized and successfully used as novel vaccine adjuvants to provide enhanced anticancer immunity. The polymers formed nanoparticles with the model vaccine, ovalbumin (OVA), and the immunostimulant toll-like receptor 3 agonist poly(I:C) (a synthetic analog of the double-stranded RNA). Higher uptake of poly(I:C) by the bone marrow-derived dendritic cells and macrophages and OVA by dendritic cells was observed when delivered using the polymer adjuvant. In vivo experiments showed that these nanoparticles remained longer in the subcutaneous injection site as compared to OVA alone and led to higher production of anti-OVA specific antibodies with prolonged immunostimulation. When OVA was combined with poly(I:C) that was either co-entrapped in the same particles or as separate particles, a comparable level of anti-OVA IgG1 antibodies and interleukin-6 (IL-6) was produced in mouse blood plasma, and a similar level of cytotoxic T lymphocyte (CTL) response in mice was stimulated as compared to OVA/Alum particles. Furthermore, tumor rejection in the mice that were vaccinated for 9 months with the formulations containing the polymer adjuvant was stronger than the other treatment groups without the polymer. Notably, the cationic polycarbonates were not associated with any adverse in vivo effects. Thus, these biodegradable polymers may be promising substitutes for aluminum-based adjuvants in vaccine formulations.


Adjuvants, Immunologic/chemistry , Polycarboxylate Cement/chemistry , Adjuvants, Immunologic/metabolism , Alum Compounds , Animals , Cancer Vaccines/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunoglobulin G/blood , Interleukin-6/blood , Mice , Mice, Inbred C57BL , Nanoparticles/chemistry , Nanoparticles/metabolism , Ovalbumin/chemistry , Ovalbumin/immunology , Poly I-C/chemistry , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Tissue Distribution
3.
Acc Chem Res ; 53(10): 2055-2067, 2020 10 20.
Article En | MEDLINE | ID: mdl-32910636

Development of vaccine technology that induces long lasting and potent adaptive immune responses is of vital importance to combat emerging pathogens and to design the next generation of cancer immunotherapies. Advanced biomaterials such as nanoparticle carriers are intensively explored to increase the efficacy and safety of vaccines and immunotherapies, based on their intrinsic potential to focus the therapeutic payload onto the relevant immune cells and to limit systemic distribution. With adaptive immune responses being primarily initiated in lymph nodes, the potency of nanoparticle vaccines in turn is tightly linked to their capacity to reach and accumulate in the lymph nodes draining the immunization site. Here, we discuss the main strategies applied to increase nanoparticle delivery to lymph nodes: (1) direct lymph node injection, (2) active cell-mediated transport through targeting of peripheral dendritic cells, and (3) exploiting passive transport through the afferent lymphatics.The intralymph nodal injection is obviously the most direct way for nanoparticles to reach lymph nodes, and multiple studies have demonstrated its capability in enhancing immunostimulant drugs' immune activation and increasing the therapeutic window. However, the requirement of using ultrasound guidance for mapping lymph nodes in patients renders intranodal administration unsuited for mass vaccination campaigns. As lymph nodes are fine structured organs with lymphocytes and chemokine gradients arrayed in a highly ordered fashion, the breakdown of such formats by the intralymph nodal injection is another concern. The exploitation of dendritic cells as live vectors for transporting nanoparticles to lymph nodes has intensively been studied both ex vivo and in vivo. While ex vivo engineering of dendritic cells in theory can achieve 100% dendritic cell-specific selectivity, a scenario impossible to be achieved in vivo, this procedure is usually laborious and complicated and entails the participation of professional staff and equipment. In addition, the poor efficiency of dendritic cell migration to the draining lymph node is another significant limitation following the injection of ex vivo cultured dendritic cells. Thus, in vivo targeting of surface receptors, particularly C-type lectin receptors, on dendritic cells by conjugating nanoparticles with antibodies or ligands is intensively studied by both academia and industry. Although such nanoparticles in vivo still face nonspecific engulfment by various phagocytes, multiple studies have shown its feasibility in targeting dendritic cells with high selectivity. Moreover, through optimizing the physicochemical properties of nanoparticles, nanoparticles can passively drain to lymph nodes carried by the interstitial flow. Compared to dendritic cell-mediated transport, passive draining is much faster and of higher efficiency. Of all such properties, size is the most important parameter as large particles (>500 nm) can only reach lymph nodes by an active cell-mediated transport. Other surface properties, such as the charge and the balance of hydrophobicity-vs-hydrophilicity, strongly influence the mobility of nanoparticles in the extracellular space. In addition, albumin, a natural fatty acid transporter, has recently been demonstrated capable of binding the amphiphiles through their lipid moiety and subsequent transporting them to lymph nodes.


Lymph Nodes/immunology , Nanoparticles/chemistry , Adaptive Immunity , Animals , Cholesterol/chemistry , Dendritic Cells/cytology , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immunity, Innate , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymph Nodes/metabolism , Mice , Nanoparticles/metabolism , Poly I-C/administration & dosage , Poly I-C/chemistry , Polymers/chemistry , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , Toll-Like Receptor 7/agonists , Toll-Like Receptor 7/metabolism
4.
Chem Commun (Camb) ; 56(53): 7273-7276, 2020 Jul 02.
Article En | MEDLINE | ID: mdl-32478368

Nanoparticle-cell-nanoparticle communication by stigmergy was demonstrated using two capped nanodevices. The first community of nanoparticles (i.e.S(RA)IFN) is loaded with 9-cis-retinoic acid and capped with interferon-γ, whereas the second community of nanoparticles (i.e.S(sulf)PIC) is loaded with sulforhodamine B and capped with poly(I:C). The uptake of S(RA)IFN by SK-BR-3 breast cancer cells enhanced the expression of TLR3 receptor facilitating the subsequent uptake of S(sulf)PIC and cell killing.


Antineoplastic Agents/metabolism , Cell Communication/drug effects , Interferon Inducers/metabolism , Nanoparticles/chemistry , Poly I-C/metabolism , Alitretinoin/chemistry , Antineoplastic Agents/chemistry , Apoptosis/drug effects , Cell Line, Tumor , Gene Expression Regulation/drug effects , Humans , Interferon Inducers/chemistry , Interferon-gamma/drug effects , Nanoparticles/metabolism , Poly I-C/chemistry , Rhodamines/chemistry , Toll-Like Receptor 3/genetics
5.
Int J Nanomedicine ; 15: 1499-1515, 2020.
Article En | MEDLINE | ID: mdl-32189965

PURPOSE: Some chemotherapeutics have been shown to induce both the release of damage-associated molecular patterns (DAMPs) and the production of type I interferon (IFN-I), leading to immunogenic cell death (ICD). However, the standard chemotherapy drug for glioma, temozolomide (TMZ), cannot induce ICD as it cannot activate IFN-I signaling. Moreover, inefficient delivery of immunostimulants across the blood-brain barrier (BBB) is the main obstacle to overcome in order to induce local immune responses in the brain. METHODS: A new oligonucleotide nanoformulation (Au@PP)/poly(I:C)) was constructed by coating gold nanoparticles (AuNPs) with methoxypolyethylene glycol (mPEG)-detachable (d)-polyethyleneimine (PEI) (Au@PP) followed by inducing the formation of electrostatic interactions with polyinosinic-polycytidylic acid (poly(I:C)). Intracranial GL261 tumor-bearing C57BL/6 mice were used to explore the therapeutic outcomes of Au@PP/poly(I:C) plus TMZ in vivo. The anti-tumor immune response in the brain induced by this treatment was analyzed by RNA sequencing and immunohistochemical analyses. RESULTS: Au@PP/poly(I:C) induced IFN-I production after endocytosis into glioma cells in vitro. Additionally, Au@PP/poly(I:C) was efficiently accumulated in the glioma tissue after intranasal administration, which allowed the nanoformulation to enter the brain while bypassing the BBB. Furthermore, Au@PP/poly(I:C) plus TMZ significantly improved the overall survival of the tumor-bearing mice compared with group TMZ only. RNA sequencing and immunohistochemical analyses revealed efficient immune response activation and T lymphocyte infiltration in the Au@PP/poly(I:C) plus TMZ group. CONCLUSION: This study demonstrates that intranasal administration of Au@PP/poly(I:C) combined with TMZ induces ICD, thereby stimulating an in situ immune response to inhibit glioma growth.


Brain Neoplasms/drug therapy , Brain Neoplasms/immunology , Glioma/drug therapy , Glioma/immunology , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/therapeutic use , Administration, Intranasal , Animals , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Delivery Systems , Female , Gold/therapeutic use , Humans , Interferon Type I/metabolism , Metal Nanoparticles/ultrastructure , Mice , Mice, Inbred C57BL , Poly I-C/chemical synthesis , Poly I-C/chemistry , Polyethylene Glycols/chemical synthesis , Polyethylene Glycols/chemistry , Polyethyleneimine/chemical synthesis , Polyethyleneimine/chemistry , Survival Analysis , T-Lymphocytes/drug effects , Temozolomide/pharmacology , Temozolomide/therapeutic use
6.
Front Immunol ; 11: 596964, 2020.
Article En | MEDLINE | ID: mdl-33391267

We designed the killed swine influenza A virus (SwIAV) H1N2 antigen (KAg) with polyriboinosinic:polyribocytidylic acid [(Poly(I:C)] adsorbed corn-derived Nano-11 particle based nanovaccine called Nano-11-KAg+Poly(I:C), and evaluated its immune correlates in maternally derived antibody (MDA)-positive pigs against a heterologous H1N1 SwIAV infection. Immunologically, in tracheobronchial lymph nodes (TBLN) detected enhanced H1N2-specific cytotoxic T-lymphocytes (CTLs) in Nano-11-KAg+Poly(I:C) vaccinates, and in commercial vaccinates detected CTLs with mainly IL-17A+ and early effector phenotypes specific to both H1N2 and H1N1 SwAIV. In commercial vaccinates, activated H1N2- and H1N1-specific IFNγ+&TNFα+, IL-17A+ and central memory T-helper/Memory cells, and in Nano-11-KAg+Poly(I:C) vaccinates H1N2-specific central memory, IFNγ+ and IFNγ+&TNFα+, and H1N1-specific IL-17A+ T-helper/Memory cells were observed. Systemically, Nano-11-KAg+Poly(I:C) vaccine augmented H1N2-specific IFNγ+ CTLs and H1N1-specific IFNγ+ T-helper/Memory cells, and commercial vaccine boosted H1N2- specific early effector CTLs and H1N1-specific IFNγ+&TNFα+ CTLs, as well as H1N2- and H1N1-specific T-helper/Memory cells with central memory, IFNγ+&TNFα+, and IL-17A+ phenotypes. Remarkably, commercial vaccine induced an increase in H1N1-specific T-helper cells in TBLN and naive T-helper cells in both TBLN and peripheral blood mononuclear cells (PBMCs), while H1N1- and H1N2-specific only T-helper cells were augmented in Nano-11-KAg+Poly(I:C) vaccinates in both TBLN and PBMCs. Furthermore, the Nano-11-KAg+Poly(I:C) vaccine stimulated robust cross-reactive IgG and secretory IgA (SIgA) responses in lungs, while the commercial vaccine elicited high levels of serum and lung IgG and serum hemagglutination inhibition (HI) titers. In conclusion, despite vast genetic difference (77% in HA gene identity) between the vaccine H1N2 and H1N1 challenge viruses in Nano-11-KAg+Poly(I:C) vaccinates, compared to over 95% identity between H1N1 of commercial vaccine and challenge viruses, the virus load and macroscopic lesions in the lungs of both types of vaccinates were comparable, but the Nano-11-KAg+Poly(I:C) vaccine cleared the virus from the nasal passage better. These data suggested the important role played by Nano-11 and Poly(I:C) in the induction of polyfunctional, cross-protective cell-mediated immunity against SwIAV in MDA-positive pigs.


Influenza A virus/immunology , Influenza Vaccines/administration & dosage , Influenza Vaccines/immunology , Nanoparticles , Orthomyxoviridae Infections/veterinary , Poly I-C , Swine Diseases/prevention & control , Vaccines, Inactivated , Animals , Antibodies, Viral/immunology , Antigens, Viral/immunology , Cross Reactions , Cytokines/metabolism , Immunity, Cellular , Immunologic Memory , Influenza Vaccines/chemistry , Nanoparticles/chemistry , Poly I-C/chemistry , Swine , Swine Diseases/immunology , Swine Diseases/virology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/metabolism , T-Lymphocytes, Helper-Inducer/immunology , T-Lymphocytes, Helper-Inducer/metabolism , Viral Load
7.
Brain Behav Immun ; 82: 160-166, 2019 11.
Article En | MEDLINE | ID: mdl-31415868

The viral mimetic polyinosinic:polycytidylic acid (poly(I:C)) is increasingly used to induce maternal immune activation (mIA) to model neurodevelopmental disorders (NDDs). Robust and reproducible phenotypes across studies are essential for the generation of models that will enhance our understanding of NDDs and enable the development of improved therapeutic strategies. However, differences in mIA-induced phenotypes using poly(I:C) have been widely observed, and this has prompted the reporting of useful and much needed methodological guidelines. Here, we perform a detailed investigation of molecular weight and endotoxin variations in poly(I:C) procured from two of the most commonly used suppliers, Sigma and InvivoGen. We demonstrate that endotoxin contamination and molecular weight differences in poly(I:C) composition lead to considerable variability in maternal IL-6 response in rats treated on gestational day (GD)15 and impact on fetal outcomes. Specifically, both endotoxin contamination and molecular weight predicted reductions in litter size on GD21. Further, molecular weight predicted a reduction in placental weight at GD21. While fetal body weight at GD21 was not affected by poly(I:C) treatment, male fetal brain weight was significantly reduced by poly(I:C), dependent on supplier. Our data are in agreement with recent reports of the importance of poly(I:C) molecular weight, and extend this work to demonstrate a key role of endotoxin on relevant phenotypic outcomes. We recommend that the source and batch numbers of poly(I:C) used should always be stated and that molecular weight variability and endotoxin contamination should be minimised for more robust mIA modelling.


Fetus/immunology , Poly I-C/chemistry , Prenatal Exposure Delayed Effects/immunology , Animals , Behavior, Animal/physiology , Cytokines/immunology , Endotoxins , Female , Infectious Disease Transmission, Vertical , Litter Size , Male , Maternal Exposure , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/immunology , Poly I-C/pharmacology , Pregnancy , Rats , Rats, Wistar , Reproducibility of Results
8.
ACS Nano ; 13(5): 4947-4959, 2019 05 28.
Article En | MEDLINE | ID: mdl-30964270

The development of an effective HIV vaccine continues to be a major health challenge since, so far, only the RV144 trial has demonstrated a modest clinical efficacy. Recently, the targeting of the 12 highly conserved protease cleavage sites (PCS1-12) has been presented as a strategy seeking to hamper the maturation and infectivity of HIV. To pursue this line of research, and because peptide antigens have low immunogenicity, we have included these peptides in engineered nanoparticles, aiming at overcoming this limitation. More specifically, we investigated whether the covalent attachment of a PCS peptide (PCS5) to polysaccharide-based nanoparticles, and their coadministration with polyinosinic:polycytidylic acid (poly(I:C)), improved the generated immune response. To this end, PCS5 was first conjugated to two different polysaccharides (chitosan and hyaluronic acid) through either a stable or a cleavable bond and then associated with an oppositely charged polymer (dextran sulfate and chitosan) and poly(I:C) to form the nanoparticles. Nanoparticles associating PCS5 by ionic interactions were used in this study as the control formulation. In vivo, all nanosystems elicited high anti-PCS5 antibodies. Nanoparticles containing PCS5 conjugated and poly(I:C) seemed to induce the strongest activation of antigen-presenting cells. Interestingly, T cell activation presented different kinetics depending on the prototype. These findings show that both the nanoparticle composition and the conjugation of the HIV peptide antigen may play an important role in the generation of humoral and cellular responses.


Antigens, Viral/immunology , HIV/immunology , Nanoparticles/chemistry , Peptides/immunology , Polysaccharides/pharmacology , Amino Acid Sequence , Animals , Antibody Formation/immunology , Chitosan/chemistry , Female , Freeze Drying , HIV/drug effects , Hyaluronic Acid/chemistry , Lymphocyte Activation/drug effects , Macrophages/drug effects , Mice, Inbred BALB C , Monocytes/drug effects , Nanoparticles/ultrastructure , Poly I-C/chemistry , Proton Magnetic Resonance Spectroscopy
9.
Vaccine ; 37(14): 1945-1953, 2019 03 28.
Article En | MEDLINE | ID: mdl-30803844

Subunit vaccines generally require adjuvants to achieve optimal immune responses. Toll-like receptor (TLR) agonists are promising immune potentiators, but rapid diffusion from the injection site reduces their local effective concentration and may cause systemic reactions. In this study, we investigated the potential of aluminum hydroxide adjuvant (AH) to adsorb the TLR3 agonist poly(I:C) and TLR9 agonist CpG and compared the effect of the combination adjuvant on the immune response with either the TLR agonists or AH alone in mice. Poly(I:C) and CpG readily adsorbed onto AH and this combination adjuvant induced a stronger IgG1 and IgG2a immune response with a significant increase of antibody avidity. The combination adjuvant enhanced antigen uptake and activation of dendritic cells in vitro. It induced an inflammatory response at the injection site similar to AH but without eosinophils which are typically observed with AH. A distinctive antigen-containing monocyte/macrophage population with an intermediate level of CD11c expression was identified in the draining lymph nodes after immunization with TLR agonists and the combination adjuvant. Injection of the combination adjuvant did not induce an increase of TNFα and CXCL10 in serum in contrast to the injection of soluble TLR agonists. These results indicate that this combination adjuvant is a promising formulation to solve some of the unmet needs of current vaccines.


Adjuvants, Immunologic/pharmacology , Aluminum Hydroxide/immunology , Antibody Affinity/immunology , Immunity, Humoral , Oligodeoxyribonucleotides/immunology , Poly I-C/immunology , Toll-Like Receptors/agonists , Adjuvants, Immunologic/chemistry , Aluminum Hydroxide/chemistry , Animals , Antigens/immunology , Cytokines/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Female , Immunoglobulin G/immunology , Mice , Oligodeoxyribonucleotides/chemistry , Poly I-C/chemistry
10.
Anal Biochem ; 564-565: 133-140, 2019 01 01.
Article En | MEDLINE | ID: mdl-29842862

Toll-like receptor 3 (TLR3), a pathogen recognition receptor of the innate immune response, recognizes and is activated by double-stranded RNA (dsRNA), which is indicative of viral exposure. A sensor design exercise was conducted, using surface plasmon resonance detection, through the examination of several immobilization approaches for TLR3 as a biorecognition element (BRE) onto a modified gold surface. To examine the TLR3-dsRNA interaction a synthetic analogue mimic, poly (I:C), was used. The interaction binding characteristics were determined and compared to literature data to establish the optimal immobilization method for the TLR3 BRE. A preliminary evaluation of the efficacy of the selected TLR3 surface as a broad-spectrum viral biosensor was also performed. Amine-coupling was found to be the most reliable method for manufacturing repeatable and consistent TLR3 BRE sensor surfaces, although this immobilization schema is not tailored to place the receptor in a spatially-specific orientation. The equilibrium dissociation constant (KD) measured for this immobilized TLR3-poly (I:C) interaction was 117 ±â€¯3.30 pM. This evaluation included a cross-reactivity study using a selection of purified E. coli and synthetic double- and single-stranded nucleic acids. The results of this design exercise and ligand binding study will inform future work towards the development of a broad-spectrum viral sensor device.


Biosensing Techniques/methods , Poly I-C/chemistry , Toll-Like Receptor 3/chemistry , Nucleic Acids , Protein Binding , Surface Plasmon Resonance
11.
J Biomed Mater Res B Appl Biomater ; 107(4): 1228-1237, 2019 05.
Article En | MEDLINE | ID: mdl-30339742

The mucosal immune system is the host's first line of defense against invasion by foreign pathogens. Gelatin nanoparticles (GNPs) are suitable carriers for the delivery of antigens via various routes of administration. In the present study, GNPs were modified with polyethyleneimine (PEI), a positively charged polymer. Then, ovalbumin (OVA) and polyinosinic:polycytidylic acid (poly(I:C)), an immunostimulant, were adsorbed onto the surface of the positively charged GNPs. We assessed whether GNPs could act as an effective mucosal vaccine that is capable of inducing both mucosal and systemic immune responses. The results showed that GNPs effectively adsorbed OVA/poly(I:C), facilitated cellular uptake by RAW 264.7 macrophage cells and murine bone marrow-derived dendritic cells (BMDCs) in vitro, and led to increased expression of the maturation markers CD80 and CD86 on BMDCs. Furthermore, GNPs induced increased secretion of proinflammatory cytokines in both RAW 264.7 and BMDCs. C57BL/6 mice that were intranasally twice-immunized with OVA/poly(I:C)-loaded GNPs produced high levels of serum OVA-specific IgG antibodies and secretory IgA in nasal and lung lavage. Spleen cells from immunized mice were collected and re-stimulated with OVA, and results showed significantly augmented production of IFN-γ, IL-4, IL-5, and IL-6 in mice that received OVA/poly(I:C)-loaded GNPs. Moreover, intranasal immunization with OVA/poly(I:C)-loaded GNPs resulted in the inhibition of EG7 tumor growth in C57BL/6 mice. Taken together, these results indicate that nasal administration of OVA/poly(I:C)-loaded GNPs elicited effective mucosal and systemic immune responses, which might be useful for further applications of antigen delivery. © 2018 Wiley Periodicals, Inc. J Biomed Mater Res Part B: Appl Biomater 107B: 1228-1237, 2019.


Adjuvants, Immunologic , Antigens , Drug Carriers , Gelatin , Immunity, Mucosal/drug effects , Immunization , Nanoparticles/chemistry , Poly I-C , Polyethyleneimine , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacokinetics , Adjuvants, Immunologic/pharmacology , Administration, Intranasal , Animals , Antigens/chemistry , Antigens/pharmacology , Bone Marrow Cells/immunology , Cytokines/immunology , Dendritic Cells/immunology , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Female , Gelatin/chemistry , Gelatin/pharmacokinetics , Gelatin/pharmacology , Mice , Nasal Absorption/drug effects , Nasal Absorption/immunology , Poly I-C/chemistry , Poly I-C/pharmacokinetics , Poly I-C/pharmacology , Polyethyleneimine/chemistry , Polyethyleneimine/pharmacokinetics , Polyethyleneimine/pharmacology , RAW 264.7 Cells
12.
Biosci Biotechnol Biochem ; 82(11): 1889-1901, 2018 Nov.
Article En | MEDLINE | ID: mdl-30079840

Polyinosinic-polycytidylic acid (PIC), a double-stranded RNA that induces innate immunity in mammals, is a candidate immunopotentiator for pharmaceuticals. The potency and adverse effects of PIC are strongly correlated with the nucleotide length, and the inability to precisely control the length in PIC production limits its practical use. Length extension during the annealing process is the major factor underlying the lack of control, but tuning the annealing conditions is insufficient to resolve this issue. In this study, we developed a novel method to produce accurate nucleotide length PIC at an industrial scale. The length extension was significantly suppressed by the assembly of multiple short polyinosinic acid molecules with one long polycytidylic acid molecule. A newly developed PIC, uPIC100-400, demonstrated a reproducible length and better storage stability than that of corresponding evenly structured PIC. Human dsRNA receptors exhibited equivalent responsiveness to uPIC100-400 and the evenly structured PIC with the same length.


Nucleic Acid Conformation , Nucleotides/chemistry , Poly I-C/chemistry , Poly I-C/chemical synthesis , Cell Line , Hot Temperature , Humans , Immunity, Innate , Poly C/chemistry , Poly I/chemistry , RNA, Double-Stranded/chemistry
13.
Chem Commun (Camb) ; 54(9): 1057-1060, 2018 Jan 25.
Article En | MEDLINE | ID: mdl-29323387

Stellated fibrous mesoporous silica nanospheres significantly improve the cellular uptake of cancer antigen and the maturation of bone marrow derived dendritic cells in vitro. Moreover, the combination of poly(I:C) with stellated fibrous MS nanospheres markedly decreases the necessary dose of poly(I:C) for anti-tumor immunity, and thus opens new opportunities for the future clinical application of poly(I:C) in cancer immunotherapy.


Immunotherapy/methods , Neoplasms/immunology , Neoplasms/therapy , Poly I-C/immunology , RNA, Double-Stranded/chemical synthesis , RNA, Double-Stranded/immunology , Silicon Dioxide/chemistry , Dendritic Cells/immunology , Humans , Particle Size , Poly I-C/administration & dosage , Poly I-C/chemistry , Porosity , RNA, Double-Stranded/chemistry , Surface Properties
14.
Proc Natl Acad Sci U S A ; 114(52): 13655-13660, 2017 12 26.
Article En | MEDLINE | ID: mdl-29229829

There is an urgent need for an effective treatment for metastatic prostate cancer (PC). Prostate tumors invariably overexpress prostate surface membrane antigen (PSMA). We designed a nonviral vector, PEI-PEG-DUPA (PPD), comprising polyethylenimine-polyethyleneglycol (PEI-PEG) tethered to the PSMA ligand, 2-[3-(1, 3-dicarboxy propyl)ureido] pentanedioic acid (DUPA), to treat PC. The purpose of PEI is to bind polyinosinic/polycytosinic acid (polyIC) and allow endosomal release, while DUPA targets PC cells. PolyIC activates multiple pathways that lead to tumor cell death and to the activation of bystander effects that harness the immune system against the tumor, attacking nontargeted neighboring tumor cells and reducing the probability of acquired resistance and disease recurrence. Targeting polyIC directly to tumor cells avoids the toxicity associated with systemic delivery. PPD selectively delivered polyIC into PSMA-overexpressing PC cells, inducing apoptosis, cytokine secretion, and the recruitment of human peripheral blood mononuclear cells (PBMCs). PSMA-overexpressing tumors in nonobese diabetic/severe combined immunodeficiency (NOD/SCID) mice with partially reconstituted immune systems were significantly shrunken following PPD/polyIC treatment, in all cases. Half of the tumors showed complete regression. PPD/polyIC invokes antitumor immunity, but unlike many immunotherapies does not need to be personalized for each patient. The potent antitumor effects of PPD/polyIC should spur its development for clinical use.


Glutamate Carboxypeptidase II/antagonists & inhibitors , Poly I-C/pharmacology , Prostatic Neoplasms/immunology , Prostatic Neoplasms/pathology , Adoptive Transfer , Animals , Antigens, Surface/genetics , Antigens, Surface/metabolism , Bystander Effect , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Disease Models, Animal , Gene Expression , Glutamate Carboxypeptidase II/genetics , Glutamate Carboxypeptidase II/metabolism , Humans , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/metabolism , Male , Mice , Poly I-C/chemistry , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Protein Binding , Tumor Burden/drug effects , Xenograft Model Antitumor Assays
15.
Acta Biomater ; 64: 401-410, 2017 12.
Article En | MEDLINE | ID: mdl-28963016

The selective activation of the immune system is a concurrent problem in the treatment of persistent diseases like viral infections (e.g. hepatitis). For the delivery of the toll-like receptor ligand poly(I:C), an immunostimulatory action was discovered earlier by hydrodynamic injection. However, this technique is not clinically transferable to human patients. A modular system where the immunoactive toll-like-receptor ligand 3 (TLR-3) poly(I:C) was incorporated into calcium phosphate nanoparticles was developed. The nanoparticles had a hydrodynamic diameter of 275nm and a zeta potential of +20mV, measured by dynamic light scattering. The diameter of the solid core was 120nm by scanning electron microscopy. In vitro, the nanoparticle uptake was investigated after 1 and 24h of incubation of THP-1 cells (macrophages) with nanoparticles by fluorescence microscopy. After intravenous injection into BALB/c and C57BL/6J mice, respectively, the in vivo uptake was especially prominent in lung and liver, 1 and 3h after the injection. Pronounced immunostimulatory effects of the nanoparticles were found in vitro with primary liver cells, i.e. Kupffer cells (KC) and liver sinusoidal endothelial cells (LSEC) from wild-type C57BL/6J mice. Thus, they represent a suitable alternative to hydrodynamic injection treatments for future vaccination concepts. STATEMENT OF SIGNIFICANCE: The selective activation of the immune system is a concurrent problem in the treatment of persistent diseases like viral infections (e.g. hepatitis). For the delivery of the toll-like receptor ligand poly(I:C), an immunostimulatory action has been discovered earlier by hydrodynamic injection. However, this technique is not clinically transferable to human patients. We have developed a modular system where poly(I:C) was incorporated into calcium phosphate nanoparticles. The uptake into relevant liver cells was studied both in vitro and in vivo. After intravenous injection into mice, the in vivo uptake was especially prominent in lung and liver, 1 and 3h after the injection. The corresponding strong immune reaction proves their high potential to turn up the immune system, e.g. against viral infections, without adverse side reactions.


Calcium Phosphates , Drug Delivery Systems/methods , Immunization/methods , Nanoparticles/chemistry , Poly I-C , Toll-Like Receptor 3/agonists , Animals , Calcium Phosphates/chemistry , Calcium Phosphates/pharmacology , Humans , Mice , Mice, Inbred BALB C , Poly I-C/chemistry , Poly I-C/pharmacology , THP-1 Cells
16.
Mol Pharm ; 14(11): 4098-4112, 2017 11 06.
Article En | MEDLINE | ID: mdl-28974092

The attenuated live vaccine strain bacille Calmette-Guérin (BCG) is currently the only available vaccine against tuberculosis (TB), but is largely ineffective against adult pulmonary TB, the most common disease form. This is in part due to BCG's ability to interfere with the host innate immune response, a feature that might be targeted to enhance the potency of this vaccine. Here, we investigated the ability of chitosan-based nanoparticles (pIC-NPs) containing polyinosinic-polycytidylic acid (poly(I:C)), an inducer of innate immunity via Toll-like receptor 3 (TLR3), to enhance the immunogenicity of BCG in mouse bone marrow derived macrophages (BMDM) in vitro. Incorporation of poly(I:C) into NPs protected it against degradation by ribonucleases and increased its uptake by mouse BMDM. Whereas soluble poly(I:C) was ineffective, pIC-NPs strongly enhanced the proinflammatory immune response of BCG-infected macrophages in a synergistic fashion, as evident by increased production of cytokines and induction of nitric oxide synthesis. Using macrophages from mice deficient in key signaling molecules involved in the pathogen recognition response, we identified combined activation of MyD88- and TRIF-dependent TLR signaling pathways to be essential for the synergistic effect between BCG and NP. Moreover, synergy was strongly dependent on the order of the two stimuli, with TLR activation by BCG functioning as the priming event for the subsequent pIC-NP stimulus, which acted through an auto-/paracrine type I interferon (IFN) feedback loop. Our results provide a foundation for a promising new approach to enhance BCG-vaccine immunogenicity by costimulation with NPs. They also contribute to a molecular understanding of the observed synergistic interaction between the pIC-NPs and BCG vaccine.


BCG Vaccine/immunology , Nanoparticles/chemistry , Poly I-C/chemistry , Animals , Immunity, Innate/physiology , Interferon Type I/metabolism , Macrophage Activation/physiology , Macrophages/metabolism , Mice , Toll-Like Receptor 3/metabolism
17.
J Control Release ; 266: 109-118, 2017 Nov 28.
Article En | MEDLINE | ID: mdl-28943194

In this study, we investigated the potential of intradermal delivery of nanoparticulate vaccines to modulate the immune response of protein antigen using hollow microneedles. Four types of nanoparticles covering a broad range of physiochemical parameters, namely poly (lactic-co-glycolic) (PLGA) nanoparticles, liposomes, mesoporous silica nanoparticles (MSNs) and gelatin nanoparticles (GNPs) were compared. The developed nanoparticles were loaded with a model antigen (ovalbumin (OVA)) with and without an adjuvant (poly(I:C)), followed by the characterization of size, zeta potential, morphology, and loading and release of antigen and adjuvant. An in-house developed hollow-microneedle applicator was used to inject nanoparticle suspensions precisely into murine skin at a depth of about 120µm. OVA/poly(I:C)-loaded nanoparticles and OVA/poly(I:C) solution elicited similarly strong total IgG and IgG1 responses. However, the co-encapsulation of OVA and poly(I:C) in nanoparticles significantly increased the IgG2a response compared to OVA/poly(I:C) solution. PLGA nanoparticles and liposomes induced stronger IgG2a responses than MSNs and GNPs, correlating with sustained release of the antigen and adjuvant and a smaller nanoparticle size. When examining cellular responses, the highest CD8+ and CD4+ T cell responses were induced by OVA/poly(I:C)-loaded liposomes. In conclusion, the applicator controlled hollow microneedle delivery is an excellent method for intradermal injection of nanoparticle vaccines, allowing selection of optimal nanoparticle formulations for humoral and cellular immune responses.


Adjuvants, Immunologic/administration & dosage , Antigens/administration & dosage , Nanoparticles/administration & dosage , Needles , Ovalbumin/administration & dosage , Poly I-C/administration & dosage , Vaccination/instrumentation , Animals , Antigens/chemistry , Drug Liberation , Female , Injections, Intradermal , Lactic Acid/administration & dosage , Lactic Acid/chemistry , Liposomes , Mice, Inbred BALB C , Mice, Inbred C57BL , Microinjections , Nanoparticles/chemistry , Ovalbumin/chemistry , Poly I-C/chemistry , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/chemistry , Polylactic Acid-Polyglycolic Acid Copolymer , Vaccination/methods
18.
J Control Release ; 265: 83-92, 2017 Nov 10.
Article En | MEDLINE | ID: mdl-28890214

Microneedles are the micrometer size devices used for the delivery of vaccines and biotherapeutics. In order to increase the vaccine efficacy and reduce the antigen dose, there is a significant need to find some adjuvants for the microneedle vaccination. In this study, zymosan, which is the cell wall preparation of Saccharomyces cerevisiae, or poly (I:C) was coated on a microneedle with inactivated influenza virus, and then immunized into BALB/c mouse to determine the immunogenicity, protection and synergetic effect between two adjuvants. As a result, the group administered with zymosan and vaccine antigen showed significantly stronger IgG response, HI titer and IgG subtypes without any adverse effects, compared to the group immunized with the vaccine antigen alone. Also, there were enhanced cellular immune responses in the group received adjuvant with vaccine antigen. In addition, they showed superior protection and lung viral reduction against lethal viral challenge. Taken together, this study confirms that zymosan can be used as an immunostimulant for microneedle vaccination.


Adjuvants, Immunologic/pharmacology , Adjuvants, Pharmaceutic/pharmacology , Influenza A Virus, H1N1 Subtype/immunology , Influenza Vaccines/administration & dosage , Needles , Poly I-C/chemistry , Zymosan/chemistry , Administration, Cutaneous , Animals , Drug Delivery Systems/methods , Female , Humans , Immunity, Cellular , Influenza Vaccines/chemistry , Mice, Inbred BALB C , Microinjections , Orthomyxoviridae Infections/immunology , Orthomyxoviridae Infections/prevention & control , Vaccination/methods , Vaccines, Inactivated/administration & dosage , Vaccines, Inactivated/chemistry
19.
ChemMedChem ; 12(10): 722-727, 2017 05 22.
Article En | MEDLINE | ID: mdl-28440596

Fully synthetic MUC1 glycopeptide antitumor vaccines have a precisely specified structure and induce a targeted immune response without suppression of the immune response when using an immunogenic carrier protein. However, tumor-associated aberrantly glycosylated MUC1 glycopeptides are endogenous structures, "self-antigens", that exhibit only low immunogenicity. To overcome this obstacle, a fully synthetic MUC1 glycopeptide antitumor vaccine was combined with poly(inosinic acid:cytidylic acid), poly(I:C), as a structurally defined Toll-like receptor 3 (TLR3)-activating adjuvant. This vaccine preparation elicited extraordinary titers of IgG antibodies which strongly bound human breast cancer cells expressing tumor-associated MUC1. Beside the humoral response, the poly(I:C) glycopeptide vaccine induced a pro-inflammatory environment, very important to overcome the immune-suppressive mechanisms, and elicited a strong cellular immune response crucial for tumor elimination.


Cancer Vaccines/immunology , Glycopeptides/immunology , Mucin-1/immunology , Poly I-C/immunology , Toll-Like Receptor 3/immunology , Vaccines, Synthetic/immunology , Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/genetics , Adjuvants, Immunologic/pharmacology , Animals , Cancer Vaccines/genetics , Cancer Vaccines/pharmacology , Dendritic Cells , Humans , Mice , Poly I-C/chemistry , Vaccines, Synthetic/genetics , Vaccines, Synthetic/pharmacology
20.
Oncotarget ; 8(15): 24046-24062, 2017 Apr 11.
Article En | MEDLINE | ID: mdl-28445962

The treatment of metastatic androgen-resistant prostate cancer remains a challenge. We describe a protein vector that selectively delivers synthetic dsRNA, polyinosinic/polycytidylic acid (polyIC), to prostate tumors by targeting prostate specific membrane antigen (PSMA), which is overexpressed on the surface of prostate cancer cells.The chimeric protein is built from the double stranded RNA (dsRNA) binding domain of PKR tethered to a single chain anti-PSMA antibody. When complexed with polyIC, the chimera demonstrates selective and efficient killing of prostate cancer cells. The treatment causes the targeted cancer cells to undergo apoptosis and to secrete toxic cytokines. In a "bystander effect", these cytokines kill neighboring cancer cells that do not necessarily overexpress PSMA, and activate immune cells that enhance the killing effect. The strong effects of the targeted polyIC are demonstrated on both 2D cell cultures and 3D tumor spheroids.


Antigens, Surface/genetics , Bystander Effect/drug effects , Bystander Effect/genetics , Genetic Vectors/genetics , Glutamate Carboxypeptidase II/genetics , RNA, Double-Stranded/genetics , Recombinant Fusion Proteins/genetics , Animals , Antigens, Surface/metabolism , Apoptosis/drug effects , Apoptosis/genetics , Cell Line, Tumor , Chemotaxis, Leukocyte/drug effects , Cytokines/biosynthesis , Disease Models, Animal , Gene Expression , Genes, Reporter , Genetic Therapy , Genetic Vectors/administration & dosage , Glutamate Carboxypeptidase II/antagonists & inhibitors , Glutamate Carboxypeptidase II/metabolism , Humans , Mice , Neoplasms/genetics , Neoplasms/mortality , Neoplasms/pathology , Neoplasms/therapy , Poly I-C/chemistry , Recombinant Fusion Proteins/metabolism , Single-Chain Antibodies/genetics , Single-Chain Antibodies/pharmacology , Spheroids, Cellular , Xenograft Model Antitumor Assays
...