Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
Add more filters










Publication year range
1.
Bioanalysis ; 16(7): 77-119, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38389403

ABSTRACT

The 17th Workshop on Recent Issues in Bioanalysis (17th WRIB) took place in Orlando, FL, USA on June 19-23, 2023. Over 1000 professionals representing pharma/biotech companies, CROs, and multiple regulatory agencies convened to actively discuss the most current topics of interest in bioanalysis. The 17th WRIB included 3 Main Workshops and 7 Specialized Workshops that together spanned 1 week to allow an exhaustive and thorough coverage of all major issues in bioanalysis of biomarkers, immunogenicity, gene therapy, cell therapy and vaccines. Moreover, in-depth workshops on "EU IVDR 2017/746 Implementation and impact for the Global Biomarker Community: How to Comply with these NEW Regulations" and on "US FDA/OSIS Remote Regulatory Assessments (RRAs)" were the special features of the 17th edition. As in previous years, WRIB continued to gather a wide diversity of international, industry opinion leaders and regulatory authority experts working on both small and large molecules as well as gene, cell therapies and vaccines to facilitate sharing and discussions focused on improving quality, increasing regulatory compliance, and achieving scientific excellence on bioanalytical issues. This 2023 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop and is aimed to provide the bioanalytical community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2023 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations on Gene Therapy, Cell therapy, Vaccines and Biotherapeutics Immunogenicity. Part 1A (Mass Spectrometry Assays and Regulated Bioanalysis/BMV), P1B (Regulatory Inputs) and Part 2 (Biomarkers, IVD/CDx, LBA and Cell-Based Assays) are published in volume 16 of Bioanalysis, issues 8 and 9 (2024), respectively.


Subject(s)
Biological Assay , Technology , Biological Assay/methods , Biomarkers/analysis , Cell- and Tissue-Based Therapy , Immunotherapy, Active
2.
PLoS One ; 18(3): e0282831, 2023.
Article in English | MEDLINE | ID: mdl-36913398

ABSTRACT

Imiquimod, a Toll-like receptor 7 (TLR7) agonist is routinely used for topical administration in basal cell carcinoma and stage zero melanoma. Similarly, the TLR agonist Bacillus Calmette-Guérin is used for the local treatment of bladder cancer and clinical trials showed treatment efficacy of intratumoral injections with TLR9 agonists. However, when administered systemically, endosomal TLR agonists cause adverse responses due to broad immune activation. Hence, strategies for targeted delivery of TLR agonists to the tumor tissue are needed to enable the widespread use of endosomal TLR agonists in the context of tumor immunotherapy. One strategy for targeted delivery of TLR agonist is their conjugation to tumor antigen-specific therapeutic antibodies. Such antibody-TLR agonist conjugates act synergistically by inducing local TLR-mediated innate immune activation which complements the anti-tumor immune mechanisms induced by the therapeutic antibody. In this study, we explored different conjugation strategies for TLR9 agonists to immunoglobulin G (IgG). We evaluated biochemical conjugation of immunostimulatory CpG oligodesoxyribonucleotides (ODN) to the HER2-specific therapeutic antibody Trastuzumab with different cross-linkers comparing stochastic with site-specific conjugation. The physiochemical make-up and biological activities of the generated Trastuzumab-ODN conjugates were characterized in vitro and demonstrated that site-specific conjugation of CpG ODN is crucial for maintaining the antigen-binding capabilities of Trastuzumab. Furthermore, site-specific conjugate was effective in promoting anti-tumor immune responses in vivo in a pseudo-metastasis mouse model with engineered human HER2-transgenic tumor cells. In this in vivo model, co-delivery of Trastuzumab and CpG ODN in form of site-specific conjugates was superior to co-injection of unconjugated Trastuzumab, CpG ODN or stochastic conjugate in promoting T cell activation and expansion. Thereby, this study highlights that site-specific conjugation of CpG ODN to therapeutic antibodies targeting tumor markers is a feasible and more reliable approach for generation of conjugates which retain and combine the functional properties of the adjuvant and the antibody.


Subject(s)
Immunoconjugates , Neoplasms , Animals , Mice , Humans , Toll-Like Receptor 9/agonists , Adjuvants, Immunologic/pharmacology , Antigens , Immunoconjugates/chemistry , Neoplasms/drug therapy , Immunoglobulin G , Trastuzumab , Oligodeoxyribonucleotides , Toll-Like Receptor 7/agonists
3.
Clin Exp Immunol ; 209(1): 64-71, 2022 07 22.
Article in English | MEDLINE | ID: mdl-35352109

ABSTRACT

Dendritic cells (DC) are crucial for the priming of T cells and thereby influence adaptive immune responses. Hence, they also represent important players in shaping anti-tumour immune responses. Cancer immunotherapy has been driven over many years by the aim to harness the T-cell stimulatory activity of these crucial antigen-presenting cells (APC). Efficient antigen delivery alone is not sufficient for full engagement of the T-cell stimulatory activity of DC and the inclusion of adjuvants triggering appropriate DC activation is essential to ensure effective anti-tumour immunity induction. While the direct engagement of DC function is a powerful tool for tumour immunotherapy, many therapeutic antibodies, such as antibodies directed against tumour-associated antigens (TAA) and immune checkpoint inhibitors (ICI) have been shown to engage DC function indirectly. The induction of anti-tumour immune responses by TAA-targeting and immune checkpoint inhibitory antibodies is thought to be integral to their therapeutic efficacy. Here, we provide an overview of the immunotherapeutic antibodies in the context of cancer immunotherapy, that has been demonstrated to directly or indirectly engage DC and discuss the current understanding of the functional mechanisms underlying anti-tumour immunity induction by these antibody therapies. In the future, the combination of therapeutic strategies that engage DC function directly and/or indirectly with strategies that allow tumour infiltrating immune effector cells to exert their anti-tumour activity in the tumour microenvironment (TME) may be key for the successful treatment of cancer patients currently not responding to immunotherapeutic antibody treatment.


Subject(s)
Dendritic Cells , Neoplasms , Antibodies , Antigens, Neoplasm , Humans , Immunotherapy , Neoplasms/therapy , T-Lymphocytes , Tumor Microenvironment
4.
J Control Release ; 297: 79-90, 2019 03 10.
Article in English | MEDLINE | ID: mdl-30659906

ABSTRACT

Tumour-specific, immuno-based therapeutic interventions can be considered as safe and effective approaches for cancer therapy. Exploitation of nano-vaccinology to intensify the cancer vaccine potency may overcome the need for administration of high vaccine doses or additional adjuvants and therefore could be a more efficient approach. Carbon nanotube (CNT) can be described as carbon sheet(s) rolled up into a cylinder that is nanometers wide and nanometers to micrometers long. Stemming from the observed capacities of CNTs to enter various types of cells via diversified mechanisms utilising energy-dependent and/or passive routes of cell uptake, the use of CNTs for the delivery of therapeutic agents has drawn increasing interests over the last decade. Here we review the previous studies that demonstrated the possible benefits of these cylindrical nano-vectors as cancer vaccine delivery systems as well as the obstacles their clinical application is facing.


Subject(s)
Adjuvants, Immunologic/chemistry , Antineoplastic Agents/chemistry , Cancer Vaccines/chemistry , Drug Carriers/chemistry , Nanotubes, Carbon/chemistry , Neoplasms/therapy , Adjuvants, Immunologic/pharmacology , Animals , Antineoplastic Agents/pharmacology , Biocompatible Materials/chemistry , Cell Line, Tumor , Cell Membrane Permeability , Drug Liberation , Humans , Molecular Targeted Therapy , Neoplasms/prevention & control , Surface Properties
5.
Nat Commun ; 9(1): 2951, 2018 07 27.
Article in English | MEDLINE | ID: mdl-30054470

ABSTRACT

Tumour-associated macrophages (TAMs) play an important role in tumour progression, which is facilitated by their ability to respond to environmental cues. Here we report, using murine models of breast cancer, that TAMs expressing fibroblast activation protein alpha (FAP) and haem oxygenase-1 (HO-1), which are also found in human breast cancer, represent a macrophage phenotype similar to that observed during the wound healing response. Importantly, the expression of a wound-like cytokine response within the tumour is clinically associated with poor prognosis in a variety of cancers. We show that co-expression of FAP and HO-1 in macrophages results from an innate early regenerative response driven by IL-6, which both directly regulates HO-1 expression and licenses FAP expression in a skin-like collagen-rich environment. We show that tumours can exploit this response to facilitate transendothelial migration and metastatic spread of the disease, which can be pharmacologically targeted using a clinically relevant HO-1 inhibitor.


Subject(s)
Breast Neoplasms/metabolism , Macrophages/metabolism , Neoplasm Metastasis , Wound Healing/physiology , Animals , Cell Line, Tumor , Cell Survival , Collagen/metabolism , Cytokines/metabolism , Endopeptidases , Female , Gelatinases/metabolism , Gene Expression Regulation, Neoplastic , Heme Oxygenase-1/metabolism , Humans , Interleukin-6/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred BALB C , Mice, Mutant Strains , Phenotype , Prognosis , Serine Endopeptidases/metabolism , Skin/metabolism , Tumor Microenvironment/physiology
6.
Cell Rep ; 24(2): 419-428, 2018 07 10.
Article in English | MEDLINE | ID: mdl-29996102

ABSTRACT

Conventional type 1 DCs (cDC1s) excel at cross-presentation of dead cell-associated antigens partly because they express DNGR-1, a receptor that recognizes exposed actin filaments on dead cells. In vitro polymerized F-actin can be used as a synthetic ligand for DNGR-1. However, cellular F-actin is decorated with actin-binding proteins, which could affect DNGR-1 recognition. Here, we demonstrate that myosin II, an F-actin-associated motor protein, greatly potentiates the binding of DNGR-1 to F-actin. Latex beads coated with F-actin and myosin II are taken up by DNGR-1+ cDC1s, and antigen associated with those beads is efficiently cross-presented to CD8+ T cells. Myosin II-deficient necrotic cells are impaired in their ability to stimulate DNGR-1 or to serve as substrates for cDC1 cross-presentation to CD8+ T cells. These results provide insights into the nature of the DNGR-1 ligand and have implications for understanding immune responses to cell-associated antigens and for vaccine design.


Subject(s)
Actins/metabolism , Antigens/metabolism , Cross-Priming/immunology , Lectins, C-Type/metabolism , Myosin Type II/metabolism , Receptors, Immunologic/metabolism , Animals , Cell Death , Cell Line , Immunization , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice, Inbred C57BL , Myosin Heavy Chains/metabolism , Phagocytosis , Protein Binding , T-Lymphocytes, Cytotoxic/immunology
7.
Clin Cancer Res ; 24(7): 1617-1628, 2018 04 01.
Article in English | MEDLINE | ID: mdl-29339440

ABSTRACT

Purpose: Unprecedented clinical outcomes have been achieved in a variety of cancers by targeting immune checkpoint molecules. This preclinical study investigates heme oxygenase-1 (HO-1), an immunosuppressive enzyme that is expressed in a wide variety of cancers, as a potential immune checkpoint target in the context of a chemotherapy-elicited antitumor immune response. We evaluate repurposing tin mesoporphyrin (SnMP), which has demonstrated safety and efficacy targeting hepatic HO in the clinic for the treatment of hyperbilirubinemia, as an immune checkpoint blockade therapy for the treatment of cancer.Experimental Design: SnMP and genetic inactivation of myeloid HO-1 were evaluated alongside 5-fluorouracil in an aggressive spontaneous murine model of breast cancer (MMTV-PyMT). Single-cell RNA sequencing analysis, tumor microarray, and clinical survival data from breast cancer patients were used to support the clinical relevance of our observations.Results: We demonstrate that SnMP inhibits immune suppression of chemotherapy-elicited CD8+ T cells by targeting myeloid HO-1 activity in the tumor microenvironment. Microarray and survival data from breast cancer patients reveal that HO-1 is a poor prognostic factor in patients receiving chemotherapy. Single-cell RNA-sequencing analysis suggests that the myeloid lineage is a significant source of HO-1 expression, and is co-expressed with the immune checkpoints PD-L1/2 in human breast tumors. In vivo, we therapeutically compare the efficacy of targeting these two pathways alongside immune-stimulating chemotherapy, and demonstrate that the efficacy of SnMP compares favorably with PD-1 blockade in preclinical models.Conclusions: SnMP could represent a novel immune checkpoint therapy, which may improve the immunological response to chemotherapy. Clin Cancer Res; 24(7); 1617-28. ©2018 AACR.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/immunology , Metalloporphyrins/pharmacology , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast Neoplasms/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Disease Models, Animal , Female , Fluorouracil/pharmacology , Heme Oxygenase-1/metabolism , Humans , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
8.
PLoS One ; 12(4): e0175712, 2017.
Article in English | MEDLINE | ID: mdl-28410380

ABSTRACT

Dendritic cells (DC) have the potential to instigate a tumour-specific immune response, but their ability to prime naïve lymphocytes depends on their activation status. Thus, for tumour immunotherapy to be effective, the provision of appropriate DC activation stimuli such as Toll-like receptor (TLR) agonists is crucial in order to overcome immunosuppression associated with the tumour microenvironment. To address this, we investigated how ovarian carcinoma (OC)-associated ascites impedes activation of DC by TLR agonists. Our results show that ascites reduces the TLR-mediated up-regulation of CD86 and partially inhibits the production of the pro-inflammatory cytokines interleukin 6 (IL-6), IL-12 and tumour necrosis factor α (TNFα) in monocyte-derived DC from healthy controls. We further observe an impaired T cell stimulatory capacity of DC upon activation with TLR agonists in the presence of ascites, indicating that their functionality is affected by the immunosuppressive factors. We identify IL-10 and prostaglandin E2 (PGE2) as the pivotal immunosuppressive components in OC-associated ascites compromising TLR-mediated DC activation. Interestingly, IL-10 is present in both ascites from patients with malignant OC and in peritoneal fluid from patients with benign ovarian conditions and both fluids have similar ability to reduce TLR-mediated DC activation. However, depletion of IL-10 from ascites revealed that the presence of paracrine IL-10 is not crucial for ascites-mediated suppression of DC activation in response to TLR activation. Unlike IL-10, PGE2 is absent from peritoneal fluid of patients with benign conditions and selectively reduces TNFα induction in response to TLR-mediated activation in the presence of OC-associated ascites. Our study highlights PGE2 as an immunosuppressive component of the malignant OC microenvironment rendering PGE2 a potentially important target for immunotherapy in OC.


Subject(s)
Dinoprostone/metabolism , Interleukin-10/metabolism , Ovarian Neoplasms/pathology , Toll-Like Receptors/metabolism , Antibodies, Neutralizing/metabolism , Ascites/metabolism , B7-2 Antigen/metabolism , Cells, Cultured , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Dinoprostone/immunology , Female , Humans , Imidazoles/toxicity , Interleukin-10/immunology , Interleukin-12/analysis , Interleukin-12/metabolism , Interleukin-6/analysis , Interleukin-6/metabolism , Lipopolysaccharides/toxicity , Lymphocyte Activation/drug effects , Monocytes/cytology , Ovarian Neoplasms/immunology , Ovarian Neoplasms/metabolism , Poly I-C/toxicity , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Toll-Like Receptors/agonists , Tumor Necrosis Factor-alpha/analysis , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
9.
Cell ; 166(4): 991-1003, 2016 Aug 11.
Article in English | MEDLINE | ID: mdl-27477514

ABSTRACT

Small immune complexes cause type III hypersensitivity reactions that frequently result in tissue injury. The responsible mechanisms, however, remain unclear and differ depending on target organs. Here, we identify a kidney-specific anatomical and functional unit, formed by resident macrophages and peritubular capillary endothelial cells, which monitors the transport of proteins and particles ranging from 20 to 700 kDa or 10 to 200 nm into the kidney interstitium. Kidney-resident macrophages detect and scavenge circulating immune complexes "pumped" into the interstitium via trans-endothelial transport and trigger a FcγRIV-dependent inflammatory response and the recruitment of monocytes and neutrophils. In addition, FcγRIV and TLR pathways synergistically "super-activate" kidney macrophages when immune complexes contain a nucleic acid. These data identify a physiological function of tissue-resident kidney macrophages and a basic mechanism by which they initiate the inflammatory response to small immune complexes in the kidney.


Subject(s)
Immune Complex Diseases/immunology , Kidney/cytology , Kidney/immunology , Macrophages/immunology , Animals , Antigen-Antibody Complex , Endothelial Cells , Macrophages/cytology , Mice, Inbred C57BL , Microscopy, Immunoelectron , Monocytes/cytology , Monocytes/immunology , Neutrophils/cytology , Neutrophils/immunology , Receptors, IgG/immunology
10.
Biomaterials ; 104: 310-22, 2016 10.
Article in English | MEDLINE | ID: mdl-27475727

ABSTRACT

Although anti-cancer immuno-based combinatorial therapeutic approaches have shown promising results, efficient tumour eradication demands further intensification of anti-tumour immune response. With the emerging field of nanovaccinology, multi-walled carbon nanotubes (MWNTs) have manifested prominent potentials as tumour antigen nanocarriers. Nevertheless, the utilization of MWNTs in co-delivering antigen along with different types of immunoadjuvants to antigen presenting cells (APCs) has not been investigated yet. We hypothesized that harnessing MWNT for concurrent delivery of cytosine-phosphate-guanine oligodeoxynucleotide (CpG) and anti-CD40 Ig (αCD40), as immunoadjuvants, along with the model antigen ovalbumin (OVA) could potentiate immune response induced against OVA-expressing tumour cells. We initially investigated the effective method to co-deliver OVA and CpG using MWNT to the APC. Covalent conjugation of OVA and CpG prior to loading onto MWNTs markedly augmented the CpG-mediated adjuvanticity, as demonstrated by the significantly increased OVA-specific T cell responses in vitro and in C57BL/6 mice. αCD40 was then included as a second immunoadjuvant to further intensify the immune response. Immune response elicited in vitro and in vivo by OVA, CpG and αCD40 was significantly potentiated by their co-incorporation onto the MWNTs. Furthermore, MWNT remarkably improved the ability of co-loaded OVA, CpG and αCD40 in inhibiting the growth of OVA-expressing B16F10 melanoma cells in subcutaneous or lung pseudo-metastatic tumour models. Therefore, this study suggests that the utilization of MWNTs for the co-delivery of tumour-derived antigen, CpG and αCD40 could be a competent approach for efficient tumours eradication.


Subject(s)
Antigen-Presenting Cells/immunology , Cancer Vaccines/administration & dosage , Nanocapsules/chemistry , Nanotubes, Carbon/chemistry , Neoplasms, Experimental/immunology , Neoplasms, Experimental/therapy , Ovalbumin/administration & dosage , Animals , Antigen-Presenting Cells/drug effects , Cell Line, Tumor , Immunotherapy/methods , Mice , Nanocapsules/administration & dosage , Neoplasms, Experimental/pathology , Treatment Outcome
11.
J Control Release ; 225: 205-16, 2016 Mar 10.
Article in English | MEDLINE | ID: mdl-26802552

ABSTRACT

Carbon nanotubes (CNTs) have shown marked capabilities in enhancing antigen delivery to antigen presenting cells. However, proper understanding of how altering the physical properties of CNTs may influence antigen uptake by antigen presenting cells, such as dendritic cells (DCs), has not been established yet. We hypothesized that altering the physical properties of multi-walled CNTs (MWNTs)-antigen conjugates, e.g. length and surface charge, can affect the internalization of MWNT-antigen by DCs, hence the induced immune response potency. For this purpose, pristine MWNTs (p-MWNTs) were exposed to various chemical reactions to modify their physical properties then conjugated to ovalbumin (OVA), a model antigen. The yielded MWNTs-OVA conjugates were long MWNT-OVA (~386nm), bearing net positive charge (5.8mV), or short MWNTs-OVA (~122nm) of increasing negative charges (-23.4, -35.8 or -39mV). Compared to the short MWNTs-OVA bearing high negative charges, short MWNT-OVA with the lowest negative charge demonstrated better cellular uptake and OVA-specific immune response both in vitro and in vivo. However, long positively-charged MWNT-OVA showed limited cellular uptake and OVA specific immune response in contrast to short MWNT-OVA displaying the least negative charge. We suggest that reduction in charge negativity of MWNT-antigen conjugate enhances cellular uptake and thus the elicited immune response intensity. Nevertheless, length of MWNT-antigen conjugate might also affect the cellular uptake and immune response potency; highlighting the importance of physical properties as a consideration in designing a MWNT-based vaccine delivery system.


Subject(s)
Drug Carriers/administration & dosage , Nanotubes, Carbon , Vaccines/administration & dosage , Animals , Antigens/administration & dosage , Antigens/chemistry , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Proliferation/drug effects , Cells, Cultured , Dendritic Cells/drug effects , Dendritic Cells/immunology , Drug Carriers/chemistry , Female , Interferon-gamma/immunology , Mice, Inbred C57BL , Mice, Knockout , Nanotubes, Carbon/chemistry , Ovalbumin/administration & dosage , Ovalbumin/chemistry , Ovalbumin/pharmacokinetics , Peptide Fragments/administration & dosage , Peptide Fragments/chemistry , Sulfhydryl Compounds/chemistry , Surface Properties , Vaccines/chemistry
12.
PLoS One ; 10(2): e0118096, 2015.
Article in English | MEDLINE | ID: mdl-25692288

ABSTRACT

Induction of potent antibody is the goal of many vaccines targeted against infections or cancer. Modern vaccine designs that use virus-like particles (VLP) have shown efficacy for prophylactic vaccination against virus-associated cancer in the clinic. Here we used plant viral particles (PVP), which are structurally analogous to VLP, coupled to a weak idiotypic (Id) tumour antigen, as a conjugate vaccine to induce antibody against a murine B-cell malignancy. The Id-PVP vaccine incorporates a natural adjuvant, the viral ssRNA, which acts via TLR7. It induced potent protective anti-Id antibody responses in an in vivo mouse model, superior to the "gold standard" Id vaccine, with prevalence of the IgG2a isotype. Combination with alum further increased antibody levels and maintained the IgG2a bias. Engagement of TLR7 in vivo was followed by secretion of IFN-α by plasmacytoid dendritic cells and by activation of splenic CD11chi conventional dendritic cells. The latter was apparent from up-regulation of co-stimulatory molecules and from secretion of a wide range of inflammatory cytokines and chemokines including the Th1-governing cytokine IL-12, in keeping with the IgG2a antibody isotype distribution. PVP conjugates are a novel cancer vaccine design, offering an attractive molecular form, similar to VLP, and providing T-cell help. In contrast to VLP, they also incorporate a safe "in-built" ssRNA adjuvant.


Subject(s)
Antibodies, Viral/immunology , Plant Viruses/immunology , Toll-Like Receptor 7/metabolism , Animals , Dendritic Cells , Female , Immunoglobulin G/immunology , Interleukin-12/metabolism , Lymphoma , Male , Mice , Mice, Inbred C57BL , Nicotiana/virology , Toll-Like Receptor 7/genetics , Virion/immunology
13.
EMBO J ; 33(6): 542-58, 2014 Mar 18.
Article in English | MEDLINE | ID: mdl-24514026

ABSTRACT

The sensing of nucleic acids by receptors of the innate immune system is a key component of antimicrobial immunity. RNA:DNA hybrids, as essential intracellular replication intermediates generated during infection, could therefore represent a class of previously uncharacterised pathogen-associated molecular patterns sensed by pattern recognition receptors. Here we establish that RNA:DNA hybrids containing viral-derived sequences efficiently induce pro-inflammatory cytokine and antiviral type I interferon production in dendritic cells. We demonstrate that MyD88-dependent signalling is essential for this cytokine response and identify TLR9 as a specific sensor of RNA:DNA hybrids. Hybrids therefore represent a novel molecular pattern sensed by the innate immune system and so could play an important role in host response to viruses and the pathogenesis of autoimmune disease.


Subject(s)
Dendritic Cells/metabolism , Immunity, Innate/immunology , Models, Immunological , Nucleic Acid Heteroduplexes/metabolism , Signal Transduction/immunology , Toll-Like Receptor 9/metabolism , Animals , Blotting, Western , Dendritic Cells/immunology , Endosomes , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , Fluorescence Polarization , Fluorescent Antibody Technique , Humans , Immunoblotting , Mice , Mice, Inbred C57BL , Myeloid Differentiation Factor 88/immunology , Nucleic Acid Heteroduplexes/immunology , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 9/immunology
14.
Article in English | MEDLINE | ID: mdl-23908972

ABSTRACT

The innate immune system has evolved endosomal and cytoplasmic receptors for the detection of viral nucleic acids as sensors for virus infection. Some of these pattern recognition receptors (PRR) detect features of viral nucleic acids that are not found in the host such as long stretches of double-stranded RNA (dsRNA) and uncapped single-stranded RNA (ssRNA) in case of Toll-like receptor (TLR) 3 and RIG-I, respectively. In contrast, TLR7/8 and TLR9 are unable to distinguish between viral and self-nucleic acids on the grounds of distinct molecular patterns. The ability of these endosomal TLR to act as PRR for viral nucleic acids seems to rely solely on the mode of access to the endolysosomal compartment in which recognition takes place. The current dogma states that self-nucleic acids do not enter the TLR-sensing compartment under normal physiological conditions. However, it is still poorly understood how dendritic cells (DC) evade activation by self-nucleic acids, in particular with regard to specific DC subsets, which are specialized in taking up material from dying cells for cross-presentation of cell-associated antigens. In this review we discuss the current understanding of how the immune system distinguishes between foreign and self-nucleic acids and point out some of the key aspects that still require further research and clarification.


Subject(s)
Endosomes/immunology , Endosomes/metabolism , Immunity, Innate , Nucleic Acids/metabolism , Receptors, Immunologic/metabolism , Viruses/immunology , Animals , Humans
15.
Cell ; 153(2): 362-75, 2013 Apr 11.
Article in English | MEDLINE | ID: mdl-23582326

ABSTRACT

The functions of Nr4a1-dependent Ly6C(low) monocytes remain enigmatic. We show that they are enriched within capillaries and scavenge microparticles from their lumenal side in a steady state. In the kidney cortex, perturbation of homeostasis by a TLR7-dependent nucleic acid "danger" signal, which may signify viral infection or local cell death, triggers Gαi-dependent intravascular retention of Ly6C(low) monocytes by the endothelium. Then, monocytes recruit neutrophils in a TLR7-dependent manner to mediate focal necrosis of endothelial cells, whereas the monocytes remove cellular debris. Prevention of Ly6C(low) monocyte development, crawling, or retention in Nr4a1(-/-), Itgal(-/-), and Tlr7(host-/-BM+/+) and Cx3cr1(-/-) mice, respectively, abolished neutrophil recruitment and endothelial killing. Prevention of neutrophil recruitment in Tlr7(host+/+BM-/-) mice or by neutrophil depletion also abolished endothelial cell necrosis. Therefore, Ly6C(low) monocytes are intravascular housekeepers that orchestrate the necrosis by neutrophils of endothelial cells that signal a local threat sensed via TLR7 followed by the in situ phagocytosis of cellular debris.


Subject(s)
Endothelial Cells/metabolism , Monitoring, Immunologic , Monocytes/immunology , Animals , Cell Adhesion Molecules/metabolism , Cell-Derived Microparticles , Endothelium, Vascular/cytology , Endothelium, Vascular/immunology , Humans , Inflammation , Intercellular Adhesion Molecule-1/metabolism , Kidney/blood supply , Kidney/metabolism , Lipopolysaccharide Receptors/metabolism , Lymphocyte Function-Associated Antigen-1/metabolism , Mice , Monocytes/metabolism , Neutrophils/immunology , Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism , Receptors, Chemokine/metabolism
16.
PLoS One ; 7(7): e40208, 2012.
Article in English | MEDLINE | ID: mdl-22808118

ABSTRACT

Antibody-antigen conjugates, which promote antigen-presentation by dendritic cells (DC) by means of targeted delivery of antigen to particular DC subsets, represent a powerful vaccination approach. To ensure immunity rather than tolerance induction the co-administration of a suitable adjuvant is paramount. However, co-administration of unlinked adjuvant cannot ensure that all cells targeted by the antibody conjugates are appropriately activated. Furthermore, antigen-presenting cells (APC) that do not present the desired antigen are equally strongly activated and could prime undesired responses against self-antigens. We, therefore, were interested in exploring targeted co-delivery of antigen and adjuvant in cis in form of antibody-antigen-adjuvant conjugates for the induction of anti-tumour immunity. In this study, we report on the assembly and characterization of conjugates consisting of DEC205-specific antibody, the model antigen ovalbumin (OVA) and CpG oligodeoxynucleotides (ODN). We show that such conjugates are more potent at inducing cytotoxic T lymphocyte (CTL) responses than control conjugates mixed with soluble CpG. However, our study also reveals that the nucleic acid moiety of such antibody-antigen-adjuvant conjugates alters their binding and uptake and allows delivery of the antigen and the adjuvant to cells partially independently of DEC205. Nevertheless, antibody-antigen-adjuvant conjugates are superior to antibody-free antigen-adjuvant conjugates in priming CTL responses and efficiently induce anti-tumour immunity in the murine B16 pseudo-metastasis model. A better understanding of the role of the antibody moiety is required to inform future conjugate vaccination strategies for efficient induction of anti-tumour responses.


Subject(s)
Adjuvants, Immunologic/administration & dosage , Antibodies/immunology , Antigens/administration & dosage , Antigens/immunology , Dendritic Cells/immunology , Drug Delivery Systems , Immunoconjugates/immunology , Amino Acid Sequence , Animals , Antigens, CD/immunology , Cross-Priming/immunology , Cytotoxicity, Immunologic , Lectins, C-Type/immunology , Mice , Mice, Inbred C57BL , Minor Histocompatibility Antigens , Molecular Sequence Data , Neoplasms/immunology , Oligodeoxyribonucleotides/immunology , Ovalbumin/immunology , Peptides/chemistry , Peptides/immunology , Receptors, Cell Surface/immunology , Solubility , T-Lymphocytes, Cytotoxic/immunology , Toll-Like Receptor 9/metabolism
17.
PLoS One ; 6(5): e19662, 2011.
Article in English | MEDLINE | ID: mdl-21637760

ABSTRACT

Dendritic cells (DCs) generated in vitro to present tumour antigens have been injected in cancer patients to boost in vivo anti-tumour immune responses. This approach to cancer immunotherapy has had limited success. For anti-tumour therapy, delivery and subsequent migration of DCs to lymph nodes leading to effective stimulation of effector T cells is thought to be essential. The ability to non-invasively monitor the fate of adoptively transferred DCs in vivo using magnetic resonance imaging (MRI) is an important clinical tool to correlate their in vivo behavior with response to treatment. Previous reports of superparamagnetic iron oxides (SPIOs) labelling of different cell types, including DCs, have indicated varying detrimental effects on cell viability, migration, differentiation and immune function. Here we describe an optimised labelling procedure using a short incubation time and low concentration of clinically used SPIO Endorem to successfully track murine DC migration in vivo using MRI in a mouse tumour model. First, intracellular labelling of bone marrow derived DCs was monitored in vitro using electron microscopy and MRI relaxometry. Second, the in vitro characterisation of SPIO labelled DCs demonstrated that viability, phenotype and functions were comparable to unlabelled DCs. Third, ex vivo SPIO labelled DCs, when injected subcutaneously, allowed for the longitudinal monitoring by MR imaging of their migration in vivo. Fourth, the SPIO DCs induced the proliferation of adoptively transferred CD4(+) T cells but, most importantly, they primed cytotoxic CD8(+) T cell responses to protect against a B16-Ova tumour challenge. Finally, using anatomical information from the MR images, the immigration of DCs was confirmed by the increase in lymph node size post-DC injection. These results demonstrate that the SPIO labelling protocol developed in this study is not detrimental for DC function in vitro and in vivo has potential clinical application in monitoring therapeutic DCs in patients with cancer.


Subject(s)
Cancer Vaccines/immunology , Dendritic Cells/immunology , Dextrans/metabolism , Melanoma, Experimental/immunology , Staining and Labeling , Vaccination , Animals , Biological Assay , Bone Marrow Cells/cytology , Cell Movement , Cell Proliferation , Cell Survival , Contrast Media/metabolism , Dendritic Cells/cytology , Dendritic Cells/ultrastructure , Lymph Nodes/metabolism , Magnetic Resonance Imaging , Magnetite Nanoparticles , Mice , Mice, Inbred C57BL , Phenotype , T-Lymphocytes/cytology
18.
Blood ; 115(10): 1949-57, 2010 Mar 11.
Article in English | MEDLINE | ID: mdl-20065291

ABSTRACT

There is a high demand for the development of adjuvants that induce cytotoxic T lymphocytes, which are crucial for the elimination of intracellular pathogens and tumor cells. Toll-like receptor (TLR) agonists are prime candidates to fulfill this role because they induce innate immune activation and promote adaptive immune responses. The successful application of the TLR7 agonist R837 for treatment of basal cell carcinoma shows the potential for exploiting this pathway in tumor immunotherapy. Imidazoquinolines like R837 and stimulatory ssRNA oligonucleotides both trigger TLR7-mediated immune activation, but little is known about their comparative ability to promote immunity induction. We investigated differences in innate immune activation and adjuvant activity between the imidazoquinoline R848 and the ssRNA TLR7 agonist polyUs21. In contrast to R848, polyUs21 induced detectable levels of intracellular interferon-alpha (IFN-alpha) in plasmacytoid dendritic cells (PDCs). In immunization studies, only polyUs21 led to robust priming of type 1 T helper cells and cytotoxic T lymphocytes, and it was more efficient in inducing antitumor immunity than R848. Notably, exogenous IFN-alpha augmented the adjuvant activity of R848, whereas depletion of PDC abrogated the adjuvanticity of polyUs21. This study, therefore, identifies sufficient IFN-alpha production by PDC as an important determinant of vaccine efficacy.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/metabolism , Interferon Type I/physiology , Membrane Glycoproteins/agonists , Toll-Like Receptor 7/agonists , Aminoquinolines/pharmacology , Aminoquinolines/therapeutic use , Animals , Antineoplastic Agents/pharmacology , Cancer Vaccines/pharmacology , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Imidazoles/pharmacology , Imidazoles/therapeutic use , Imiquimod , Interferon Type I/metabolism , Melanoma, Experimental/immunology , Melanoma, Experimental/metabolism , Melanoma, Experimental/therapy , Mice , Mice, Inbred C57BL , Mice, Knockout , Quinolines/pharmacology , RNA/pharmacology , Receptor, Interferon alpha-beta/genetics , Tumor Cells, Cultured
19.
Immunol Lett ; 128(1): 17-20, 2010 Jan 18.
Article in English | MEDLINE | ID: mdl-19808054

ABSTRACT

Innate recognition of viruses is mediated by a panel of intracellular receptors that sense viral genomic nucleic acids and/or replication intermediates. Recognition of these viral signatures takes place in endosomal compartments of specialised cell types via a particular set of Toll-like receptors upon uptake of viral particles or material from infected dying cells. In addition, ubiquitously expressed nucleic acid-sensing molecules in the cytoplasm allow infected cells to detect virus replication and to alert neighbouring cells. This article gives an overview over the different viral nucleic acid sensors and their contribution to the orchestration of anti-viral immune responses.


Subject(s)
Immunity, Innate , Toll-Like Receptors/immunology , Virus Diseases/immunology , Viruses/immunology , Animals , Cytoplasm/immunology , Cytoplasm/virology , DNA, Viral/metabolism , Endosomes/immunology , Endosomes/virology , Host-Pathogen Interactions , Humans , Interferons/immunology , RNA, Viral/metabolism , Viruses/pathogenicity
20.
Int Immunol ; 21(7): 871-9, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19505890

ABSTRACT

Dendritic cells (DC) are key players in the initiation and modulation of adaptive immune responses due to their ability to acquire and present antigen and stimulate T cells. For the induction of effector T cell functions, antigen must be presented by activated DC. In this study, we have compared uptake of antigen by mouse DC in the presence of different Toll-like receptor (TLR) agonists, which are potent inducers of DC activation. Here we show that the reduction in uptake of soluble antigen in the presence of the viral double-stranded RNA (dsRNA) analogues polyinosinic-polycytidylic acid and Ampligen is independent of TLR-mediated DC activation. A reduction in antigen uptake by bone marrow-derived and splenic DC was also observed in response to other RNA homopolymers such as polyinosinic and polyguanylic acids, which are known inhibitors of scavenger receptor-mediated endocytosis. Pinocytosis and mannose receptor-mediated uptake of soluble antigen were not affected by any of the tested nucleic acids. The reduction in antigen uptake by dsRNA did not negatively influence the T cell stimulating properties of the DC. In summary, we conclude that the decrease in antigen endocytosis observed in the presence of a variety of TLR agonists is independent of TLR signalling and is caused by competition for specific surface receptors that are involved in the uptake of these TLR agonists and the antigen.


Subject(s)
Adjuvants, Immunologic/pharmacology , Dendritic Cells/drug effects , Interferon Inducers/pharmacology , Poly I-C/pharmacology , RNA, Double-Stranded/pharmacology , Toll-Like Receptors/agonists , Animals , Antigens/immunology , Dendritic Cells/immunology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lipopolysaccharides/pharmacology , Lymphocyte Activation/drug effects , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Oligodeoxyribonucleotides/pharmacology , Pinocytosis/drug effects , Pinocytosis/immunology , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Toll-Like Receptors/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...