Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 167
Filter
Add more filters

Publication year range
1.
Immunity ; 42(5): 850-63, 2015 May 19.
Article in English | MEDLINE | ID: mdl-25979419

ABSTRACT

The molecular mechanisms regulating antigen translocation into the cytosol for cross-presentation are under controversial debate, mainly because direct data is lacking. Here, we have provided direct evidence that the activity of the endoplasmic reticulum (ER) translocon protein Sec61 is essential for endosome-to-cytosol translocation. We generated a Sec61-specific intrabody, a crucial tool that trapped Sec61 in the ER and prevented its recruitment into endosomes without influencing Sec61 activity and antigen presentation in the ER. Expression of this ER intrabody inhibited antigen translocation and cross-presentation, demonstrating that endosomal Sec61 indeed mediates antigen transport across endosomal membranes. Moreover, we showed that the recruitment of Sec61 toward endosomes, and hence antigen translocation and cross-presentation, is dependent on dendritic cell activation by Toll-like receptor (TLR) ligands. These data shed light on a long-lasting question regarding antigen cross-presentation and point out a role of the ER-associated degradation machinery in compartments distinct from the ER.


Subject(s)
Antigens/metabolism , CD8-Positive T-Lymphocytes , Cross-Priming/immunology , Cytosol/metabolism , Endosomes/metabolism , Membrane Proteins/metabolism , Models, Biological , Animals , Antigens/immunology , Cell Line , Cytosol/immunology , Membrane Proteins/chemistry , Mice , Protein Transport , SEC Translocation Channels
2.
Cancer Immunol Immunother ; 72(7): 2405-2422, 2023 Jul.
Article in English | MEDLINE | ID: mdl-36997666

ABSTRACT

Metastases remain the leading cause of cancer-related death worldwide. Therefore, improving the treatment efficacy against such tumors is essential to enhance patient survival. AU-011 (belzupacap sarotalocan) is a new virus-like drug conjugate which is currently in clinical development for the treatment of small choroidal melanoma and high-risk indeterminate lesions in the eye. Upon light activation, AU-011 induces rapid necrotic cell death which is pro-inflammatory and pro-immunogenic, resulting in an anti-tumor immune response. As AU-011 is known to induce systemic anti-tumor immune responses, we investigated whether this combination therapy would also be effective against distant, untreated tumors, as a model for treating local and distant tumors by abscopal immune effects. We compared the efficacy of combining AU-011 with several different checkpoint blockade antibodies to identify optimal treatment regimens in an in vivo tumor model. We show that AU-011 induces immunogenic cell death through the release and exposure of damage-associated molecular patterns (DAMPs), resulting in the maturation of dendritic cells in vitro. Furthermore, we show that AU-011 accumulates in MC38 tumors over time and that ICI enhances the efficacy of AU-011 against established tumors in mice, resulting in complete responses for specific combinations in all treated animals bearing a single MC38 tumor. Finally, we show that AU-011 and anti-PD-L1/anti-LAG-3 antibody treatment was an optimal combination in an abscopal model, inducing complete responses in approximately 75% of animals. Our data show the feasibility of combining AU-011 with PD-L1 and LAG-3 antibodies for the treatment of primary and distant tumors.


Subject(s)
Immune Checkpoint Inhibitors , Melanoma , Animals , Mice , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Disease Models, Animal , Melanoma/drug therapy , Combined Modality Therapy , Photosensitizing Agents , Cell Line, Tumor
3.
Nat Immunol ; 12(1): 45-53, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21151101

ABSTRACT

Cytotoxic T lymphocytes (CTLs) recognize peptides presented by HLA class I molecules on the cell surface. The C terminus of these CTL epitopes is considered to be produced by the proteasome. Here we demonstrate that the cytosolic endopeptidases nardilysin and thimet oligopeptidase (TOP) complemented proteasome activity. Nardilysin and TOP were required, either together or alone, for the generation of a tumor-specific CTL epitope from PRAME, an immunodominant CTL epitope from Epstein-Barr virus protein EBNA3C, and a clinically important epitope from the melanoma protein MART-1. TOP functioned as C-terminal trimming peptidase in antigen processing, and nardilysin contributed to both the C-terminal and N-terminal generation of CTL epitopes. By broadening the antigenic peptide repertoire, nardilysin and TOP strengthen the immune defense against intracellular pathogens and cancer.


Subject(s)
Antigens, Neoplasm/metabolism , Epitopes, T-Lymphocyte/metabolism , Metalloendopeptidases/metabolism , T-Lymphocytes, Cytotoxic/metabolism , Antigen Presentation/genetics , Antigens, Neoplasm/chemistry , Antigens, Neoplasm/immunology , Cytotoxicity, Immunologic/genetics , Epitopes, T-Lymphocyte/chemistry , Epitopes, T-Lymphocyte/immunology , HLA-A3 Antigen/metabolism , Humans , K562 Cells , Metalloendopeptidases/genetics , Metalloendopeptidases/immunology , Peptide Fragments/chemistry , Peptide Fragments/immunology , Peptide Fragments/metabolism , Proteasome Endopeptidase Complex/metabolism , Protein Binding , RNA, Small Interfering/genetics , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/pathology , Transgenes/genetics
4.
Eur J Immunol ; 51(4): 835-847, 2021 04.
Article in English | MEDLINE | ID: mdl-33349928

ABSTRACT

Autophagy has been reported to be involved in supporting antigen cross-presentation by dendritic cells (DCs). We have shown that DCs have the ability to store antigen for a prolonged time in endolysosomal compartments and thereby sustain MHCI antigen cross-presentation to CD8+ T cells. In the current study, we investigated the role of autophagy in long-term antigen presentation. We show that the autophagy machinery has a negative impact on storage of antigen in DCs. Atg5-/- DCs which are deficient in autophagy or DCs treated with common autophagy inhibitors showed enhanced antigen storage and antigen cross-presentation. This augmented antigen cross-presentation effect is independent of altered proteasome enzyme activity or MHCI surface expression on DCs. We visualized that the storage compartments are in close proximity to LC3 positive autophagosomes. Our results indicate that autophagosomes disrupt antigen storage in DCs and thereby regulate long-term MHCI cross-presentation.


Subject(s)
Antigen Presentation/immunology , Autophagy/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Animals , Antigen Presentation/drug effects , Autophagosomes/immunology , Autophagosomes/metabolism , Autophagy/drug effects , Autophagy-Related Protein 5/genetics , Autophagy-Related Protein 5/immunology , Autophagy-Related Protein 5/metabolism , CD8-Positive T-Lymphocytes/immunology , Cell Line , Cross-Priming/drug effects , Dendritic Cells/cytology , Dendritic Cells/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Microtubule-Associated Proteins/immunology , Microtubule-Associated Proteins/metabolism , Wortmannin/pharmacology
5.
Eur J Immunol ; 51(8): 1911-1920, 2021 08.
Article in English | MEDLINE | ID: mdl-34106465

ABSTRACT

Immunotherapy targeting the Programmed Death (PD-1) receptor/ligand (L) "checkpoint" rapidly gains ground in the treatment of many cancer types. To increase treatment scope and efficacy, predictive biomarkers and rational selection of co-treatments are required. To meet these demands, we must understand PD-1 function in detail. We here outline recent insights into the regulation of the CD8+ T cell response by PD-1. The prevailing view has been that blockade of PD-1/ligand (L) interaction "reinvigorates" cytotoxic T lymphocytes (CTL) that were rendered dysfunctional in the tumor microenvironment (TME). However, this review stresses that tumors continuously communicate with adjacent draining lymph nodes (LNs) and that the PD-1 checkpoint also operates during T cell priming. We clarify the role of the PD-(L)1 system at the T cell/DC interface, where it regulates T cell receptor (TCR) signaling and CD28 costimulation and thus controls activation of tumor-specific T cells. We also highlight the importance of CD4+ T cell help during priming, which allows DCs to provide other costimulatory and cytokine signals required for optimal CTL differentiation and likely avoidance of a dysfunctional state. Therefore, we pose that PD-(L)1 blockade should exploit LN function and be combined with "help" signals to optimize CTL efficacy.


Subject(s)
B7-H1 Antigen/antagonists & inhibitors , Immunotherapy/methods , Neoplasms/drug therapy , Neoplasms/immunology , Programmed Cell Death 1 Receptor/antagonists & inhibitors , Animals , CD4-Positive T-Lymphocytes/immunology , Humans , Immune Checkpoint Inhibitors/pharmacology , T-Lymphocytes, Cytotoxic/immunology
6.
J Nanobiotechnology ; 20(1): 252, 2022 Jun 03.
Article in English | MEDLINE | ID: mdl-35658868

ABSTRACT

Extracellular vesicles (EVs) are promising drug carriers of photosensitizers for photodynamic therapy (PDT) in cancer treatment, due to their ability to circulate in blood and enter cells efficiently. The therapeutic potential of EVs has been suggested to depend on the type and physiological state of their cell of origin. However, the effects of deriving EVs from various cells in different physiological states on their antitumor capacity are rarely evaluated. In the present study, we compared the antitumor efficacy of EV-mediated PDT by incorporating the photosensitizer Zinc Phthalocyanine (ZnPc) into EVs from multiple cells sources. ZnPc was incorporated by a direct incubation strategy into EVs derived from immune cells (M1-like macrophages and M2-like macrophages), cancer cells (B16F10 melanoma cancer cells) and external sources (milk). Our data show that all EVs are suitable carriers for ZnPc and enable efficient PDT in vitro in co-culture models and in vivo. We observed that EV-mediated PDT initiates immunogenic cell death through the release and exposure of damage associated molecular patterns (DAMPs) on cancer cells, which subsequently induced dendritic cell (DC) maturation. Importantly, of all ZnPc-EVs tested, in absence of light only M1-ZnPc displayed toxicity to MC38, but not to DC, in monoculture and in co-culture, indicating specificity for cancer over immune cells. In MC38 tumor-bearing mice, only M1-ZnPc induced a tumor growth delay compared to control in absence of light. Interestingly, M1- but not M2-mediated PDT, induced complete responses against MC38 tumors in murine models (100% versus 38% of cases, respectively), with survival of all animals up to at least 60 days post inoculation. Finally, we show that all cured animals are protected from a rechallenge with MC38 cells, suggesting the induction of immunological memory after EV-mediated PDT. Together, our data show the importance of the cell type from which the EVs are obtained and highlight the impact of the immunological state of these cells on the antitumor efficacy of EV-mediated PDT.


Subject(s)
Colonic Neoplasms , Extracellular Vesicles , Photochemotherapy , Animals , Cell Line, Tumor , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Extracellular Vesicles/metabolism , Immunologic Memory , Indoles/pharmacology , Mice , Photosensitizing Agents/pharmacology , Photosensitizing Agents/therapeutic use
7.
Mikrochim Acta ; 189(10): 368, 2022 09 03.
Article in English | MEDLINE | ID: mdl-36057018

ABSTRACT

Upconversion nanoparticles (UCNPs) represent a group of NPs that can convert near-infrared (NIR) light into ultraviolet and visible light, thus possess deep tissue penetration power with less background fluorescence noise interference, and do not induce damage to biological tissues. Due to their unique optical properties and possibility for surface modification, UCNPs can be exploited for concomitant antigen delivery into dendritic cells (DCs) and monitoring by molecular imaging. In this study, we focus on the development of a nano-delivery platform targeting DCs for immunotherapy and simultaneous imaging. OVA 254-267 (OVA24) peptide antigen, harboring a CD8 T cell epitope, and Pam3CysSerLys4 (Pam3CSK4) adjuvant were chemically linked to the surface of UCNPs by amide condensation to stimulate DC maturation and antigen presentation. The OVA24-Pam3CSK4-UCNPs were thoroughly characterized and showed a homogeneous morphology and surface electronegativity, which promoted a good dispersion of the NPs. In vitro experiments demonstrated that OVA24-Pam3CSK4-UCNPs induced a strong immune response, including DC maturation, T cell activation, and proliferation, as well as interferon gamma (IFN-γ) production. In vivo, highly sensitive upconversion luminescence (UCL) imaging of OVA24-Pam3CSK4-UCNPs allowed tracking of UCNPs from the periphery to lymph nodes. In summary, OVA24-Pam3CSK4-UCNPs represent an effective tool for DC-based immunotherapy.


Subject(s)
Nanoparticles , Dendritic Cells , Light , Luminescence , Molecular Imaging , Nanoparticles/chemistry
8.
Immunology ; 164(3): 494-506, 2021 11.
Article in English | MEDLINE | ID: mdl-34110622

ABSTRACT

An exclusive feature of dendritic cells (DCs) is their capacity to present exogenous antigens by MHC class I molecules, called cross-presentation. Here, we show that protein antigen can be conserved in mature murine DCs for several days in a lysosome-like storage compartment, distinct from MHC class II and early endosomal compartments, as an internal source for the supply of MHC class I ligands. Using two different uptake routes via Fcγ receptors and C-type lectin receptors, we could show that antigens were routed towards the same endolysosomal compartments after 48 h. The antigen-containing compartments lacked co-expression of molecules involved in MHC class I processing and presentation including TAP and proteasome subunits as shown by single-cell imaging flow cytometry. Moreover, we observed the absence of cathepsin S but selective co-localization of active cathepsin X with protein antigen in the storage compartments. This indicates cathepsin S-independent antigen degradation and a novel but yet undefined role for cathepsin X in antigen processing and cross-presentation by DCs. In summary, our data suggest that these antigen-containing compartments in DCs can conserve protein antigens from different uptake routes and contribute to long-lasting antigen cross-presentation.


Subject(s)
Antigens/metabolism , Cross-Priming , Dendritic Cells/immunology , Lectins, C-Type/metabolism , Receptors, IgG/metabolism , Animals , Antigen Presentation , Antigens/immunology , Cathepsins/metabolism , Dendritic Cells/metabolism , Dendritic Cells/ultrastructure , Endosomes/immunology , Endosomes/metabolism , Endosomes/ultrastructure , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Lysosomes/immunology , Lysosomes/metabolism , Lysosomes/ultrastructure , Mice , Microscopy, Electron, Transmission , Models, Animal , NIH 3T3 Cells , Primary Cell Culture
9.
Chembiochem ; 22(2): 434-440, 2021 01 15.
Article in English | MEDLINE | ID: mdl-32864819

ABSTRACT

Mannose-6-phosphate (M6P) is recognized by the mannose-6-phosphate receptor and plays an important role in the transport of cargo to the endosomes, making it an attractive tool to improve endosomal trafficking of vaccines. We describe herein the assembly of peptide antigen conjugates carrying clusters of mannose-6-C-phosphonates (M6Po). The M6Po's are stable M6P mimics that are resistant to cleavage of the phosphate group by endogenous phosphatases. Two different strategies for the incorporation of the M6Po clusters in the conjugate have been developed: the first relies on a "post-assembly" click approach employing an M6Po bearing an alkyne functionality; the second hinges on an M6Po C-glycoside amino acid building block that can be used in solid-phase peptide synthesis. The generated conjugates were further equipped with a TLR7 ligand to stimulate dendritic cell (DC) maturation. While antigen presentation is hindered by the presence of the M6Po clusters, the incorporation of the M6Po clusters leads to increased activation of DCs, thus demonstrating their potential in improving vaccine adjuvanticity by intraendosomally active TLR ligands.


Subject(s)
Antigens/metabolism , Mannosephosphates/metabolism , Peptides/metabolism , Toll-Like Receptors/metabolism , Antigens/chemistry , Humans , Ligands , Mannosephosphates/chemistry , Molecular Structure , Peptides/chemistry , Toll-Like Receptors/chemistry
10.
Chembiochem ; 22(7): 1215-1222, 2021 04 06.
Article in English | MEDLINE | ID: mdl-33180981

ABSTRACT

Synthetic vaccines, based on antigenic peptides that comprise MHC-I and MHC-II T-cell epitopes expressed by tumors, show great promise for the immunotherapy of cancer. For optimal immunogenicity, the synthetic peptides (SPs) should be adjuvanted with suitable immunostimulatory additives. Previously, we have shown that improved immunogenicity in vivo is obtained with vaccine modalities in which an SP is covalently connected to an adjuvanting moiety, typically a ligand to Toll-like receptor 2 (TLR2). SPs were covalently attached to UPam, which is a derivative of the classic TLR2 ligand Pam3 CysSK4 . A disadvantage of the triply palmitoylated UPam is its high lipophilicity, which precludes universal adoption of this adjuvant for covalent modification of various antigenic peptides as it renders the synthetic vaccine insoluble in several cases. Here, we report a novel conjugatable TLR2 ligand, mini-UPam, which contains only one palmitoyl chain, rather than three, and therefore has less impact on the solubility and other physicochemical properties of a synthetic peptide. In this study, we used SPs that contain the clinically relevant neoepitopes identified in a melanoma patient who completely recovered after T-cell therapy. Homogeneous mini-UPam-SP conjugates have been prepared in good yields by stepwise solid-phase synthesis that employed a mini-UPam building block pre-prepared in solution and the standard set of Fmoc-amino acids. The immunogenicity of the novel mini-UPam-SP conjugates was demonstrated by using the cancer patient's T-cells.


Subject(s)
Antigens, Neoplasm/chemistry , Cancer Vaccines/immunology , Ligands , Toll-Like Receptor 2/chemistry , Vaccines, Synthetic/immunology , Antigens, Neoplasm/immunology , Cancer Vaccines/chemistry , Cell Line , Dendritic Cells/cytology , Dendritic Cells/metabolism , Drug Design , Humans , Interleukin-8/metabolism , Lipopeptides/chemical synthesis , Lipopeptides/chemistry , Lipopeptides/immunology , Lipoylation , Lymphocyte Activation , Toll-Like Receptor 2/metabolism , Vaccines, Synthetic/chemistry
11.
Int J Mol Sci ; 22(17)2021 Aug 30.
Article in English | MEDLINE | ID: mdl-34502307

ABSTRACT

Tumor-draining lymph nodes play a paradoxical role in cancer. Surgeons often resect these sentinel lymph nodes to determine metastatic spread, thereby enabling prognosis and treatment. However, lymph nodes are vital organs for the orchestration of immune responses, due to the close encounters of dedicated immune cells. In view of the success of immunotherapy, the removal of tumor-draining lymph nodes needs to be re-evaluated and viewed in a different light. Recently, an important role for tumor-draining lymph nodes has been proposed in the immunotherapy of cancer. This new insight can change the use of immune checkpoint therapy, particularly with respect to the use in neoadjuvant settings in which lymph nodes are still operational.


Subject(s)
Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy/methods , Lymphocytes, Tumor-Infiltrating/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Sentinel Lymph Node/immunology , Animals , Humans , Neoadjuvant Therapy , Neoplasms/pathology
12.
J Immunol ; 201(12): 3741-3749, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30397036

ABSTRACT

Therapy with tumor-specific Abs is common in the clinic but has limited success against solid malignancies. We aimed at improving the efficacy of this therapy by combining a tumor-specific Ab with immune-activating compounds. In this study, we demonstrate in the aggressive B16F10 mouse melanoma model that concomitant application of the anti-TRP1 Ab (clone TA99) with TLR3-7/8 or -9 ligands, and IL-2 strongly enhanced tumor control in a therapeutic setting. Depletion of NK cells, macrophages, or CD8+ T cells all mitigated the therapeutic response, showing a coordinated immune rejection by innate and adaptive immune cells. FcγRs were essential for the therapeutic effect, with a dominant role for FcγRI and a minor role for FcγRIII and FcγRIV. FcγR expression on NK cells and granulocytes was dispensable, indicating that other tumoricidal functions of NK cells were involved and implicating that FcγRI, -III, and -IV exerted their activity on macrophages. Indeed, F4/80+Ly-6C+ inflammatory macrophages in the tumor microenvironment displayed high levels of these receptors. Whereas administration of the anti-TRP1 Ab alone reduced the frequency of these macrophages, the combination with a TLR agonist retained these cells in the tumor microenvironment. Thus, the addition of innate stimulatory compounds, such as TLR ligands, to tumor-specific Ab therapy could greatly enhance its efficacy in solid cancers via optimal exploitation of FcγRs.


Subject(s)
Antibodies/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Immunotherapy/methods , Interleukin-2/therapeutic use , Killer Cells, Natural/immunology , Macrophages/immunology , Melanoma/therapy , Receptors, IgG/metabolism , Animals , Antigens, Neoplasm/immunology , Disease Models, Animal , Female , Gene Expression Regulation, Neoplastic , Humans , Immunization , Male , Melanoma/immunology , Melanoma, Experimental , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Oxidoreductases/immunology , Receptors, IgG/genetics , Toll-Like Receptors/agonists
13.
J Immunol ; 201(1): 87-97, 2018 07 01.
Article in English | MEDLINE | ID: mdl-29752315

ABSTRACT

Enhancing T cell responses against both viral and tumor Ags requires efficient costimulation and directed delivery of peptide Ags into APCs. Long peptide vaccines are considered favorable vaccine moieties from a clinical perspective, as they can harbor more than one immunogenic epitope enabling treatment of a broader target population. In addition, longer peptides are not extracellularly loaded on MHC class I; rather, they require intracellular processing and will thereby be presented to T cells mainly by professional APCs, thereby avoiding the risk of tolerance induction. The drawback of peptide vaccines regardless of peptide length is that naked peptides are not actively targeted to and taken up by APCs, and the standard nonconjugated adjuvant-peptide mixtures do not ensure cotargeting of the two to the same APC. We have identified a tetanus toxin-derived B cell epitope that can mediate the formation of immune complexes in the presence of circulating Abs. In this study, we show that these immune complexes improve both Ag uptake by APCs (blood monocytes and CD1c+ dendritic cells) and consequently improve CD8+ T cell recall responses in a human ex vivo blood loop system. The uptake of the peptide conjugate by blood monocytes is dependent on Abs and the complement component C1q. We envision that this strategy can be used to facilitate active uptake of Ags into APCs to improve T cell responses against pathogens or cancer.


Subject(s)
Adjuvants, Immunologic/pharmacology , Antigen Presentation/immunology , Antigen-Antibody Complex/immunology , CD8-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Epitopes, B-Lymphocyte/immunology , Tetanus Toxoid/immunology , Antigens/immunology , Complement C1q/immunology , Epitopes, T-Lymphocyte/immunology , Humans , Interferon-gamma/immunology , Monocytes/immunology
14.
J Immunol ; 200(8): 2615-2626, 2018 04 15.
Article in English | MEDLINE | ID: mdl-29523656

ABSTRACT

By their interaction with IgG immune complexes, FcγR and complement link innate and adaptive immunity, showing functional redundancy. In complement-deficient mice, IgG downstream effector functions are often impaired, as well as adaptive immunity. Based on a variety of model systems using FcγR-knockout mice, it has been concluded that FcγRs are also key regulators of innate and adaptive immunity; however, several of the model systems underpinning these conclusions suffer from flawed experimental design. To address this issue, we generated a novel mouse model deficient for all FcγRs (FcγRI/II/III/IV-/- mice). These mice displayed normal development and lymphoid and myeloid ontogeny. Although IgG effector pathways were impaired, adaptive immune responses to a variety of challenges, including bacterial infection and IgG immune complexes, were not. Like FcγRIIb-deficient mice, FcγRI/II/III/IV-/- mice developed higher Ab titers but no autoantibodies. These observations indicate a redundant role for activating FcγRs in the modulation of the adaptive immune response in vivo. We conclude that FcγRs are downstream IgG effector molecules with a restricted role in the ontogeny and maintenance of the immune system, as well as the regulation of adaptive immunity.

15.
Molecules ; 25(9)2020 May 08.
Article in English | MEDLINE | ID: mdl-32397328

ABSTRACT

(1) Background: doxorubicin is a potent chemotherapeutic agent, but it has limitations regarding its side effects and therapy resistance. Hydrogels potentially deal with these problems, but several characterizations need to be optimized to better understand how hydrogel assisted chemotherapy works. Poloxamer 407 (P407) hydrogels were mixed with doxorubicin and physico-chemical, biological, and pharmacological characterizations were considered. (2) Methods: hydrogels were prepared by mixing P407 in PBS at 4 °C. Doxorubicin was added upon solutions became clear. Time-to-gelation, hydrogel morphology, and micelles were studied first. The effects of P407-doxorubicin were evaluated on MC-38 colon cancer cells. Furthermore, doxorubicin release was assessed and contrasted with non-invasive in vivo whole body fluorescence imaging. (3) Results: 25% P407 had favorable gelation properties with pore sizes of 30-180 µm. P407 micelles were approximately 5 nm in size. Doxorubicin was fully released in vitro from 25% P407 hydrogel within 120 h. Furthermore, P407 micelles strongly enhanced the anti-neoplastic effects of doxorubicin on MC-38 cells. In vivo fluorescence imaging revealed that hydrogels retained fluorescence signals at the injection site for 168 h. (4) Conclusions: non-invasive imaging showed how P407 gels retained drug at the injection site. Doxorubicin P407 micelles strongly enhanced the anti-tumor effects.


Subject(s)
Antineoplastic Agents , Colonic Neoplasms , Doxorubicin , Drug Carriers , Hydrogels , Optical Imaging , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Colonic Neoplasms/diagnostic imaging , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Carriers/chemistry , Drug Carriers/pharmacology , Humans , Hydrogels/chemistry , Hydrogels/pharmacology , Mice , Micelles
16.
Int J Cancer ; 144(2): 345-354, 2019 01 15.
Article in English | MEDLINE | ID: mdl-30259976

ABSTRACT

Immunomodulatory antibodies blocking interactions of coinhibitory receptors to their ligands such as CTLA-4, PD1 and PD-L1 on immune cells have shown impressive therapeutic efficacy in clinical studies. The therapeutic effect of these antibodies is mainly mediated by reactivating antitumor T cell immune responses. Detailed analysis of anti-CTLA4 antibody therapy revealed that an optimal therapeutic efficacy also requires binding to Fc receptors for IgG, FcγR, mediating depletion of intratumoral regulatory T cells. Here, we investigated the role of Fc binding in anti-PD-L1 antibody therapy in the MC38 C57BL/6 and CT26 BALB/c colon adenocarcinoma tumor models. In the MC38 tumor model, all IgG subclasses anti-PD-L1 showed similar therapeutic efficacy when compared to each other in either wild-type mice or in mice deficient for all FcγR. In contrast, in the CT26 tumor model, anti-PD-L1 mIgG2a, the IgG subclass with the highest affinity for activating FcγR, showed stronger therapeutic efficacy than other IgG subclasses. This was associated with a reduction of a myeloid cell subset with high expression of PD-L1 in the tumor microenvironment. This subclass preference for mIgG2a was lost in C57BL/6 × BALB/c F1 mice, indicating that the genetic background of the host may determine the additional clinical benefit of the high affinity antibody subclasses. Based on these data, we conclude that FcγR are not crucial for anti-PD-L1 antibody therapy but might play a role in some tumor models.


Subject(s)
Adenocarcinoma , Antineoplastic Agents, Immunological/pharmacology , B7-H1 Antigen/antagonists & inhibitors , Colonic Neoplasms , Receptors, IgG , Animals , Antibodies, Monoclonal , Disease Models, Animal , Immunotherapy/methods , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL
17.
Eur J Immunol ; 48(7): 1164-1173, 2018 07.
Article in English | MEDLINE | ID: mdl-29676785

ABSTRACT

An exclusive feature of dendritic cells (DCs) is their ability to cross-present exogenous antigens in MHC class I molecules. We analyzed the fate of protein antigen in antigen presenting cell (APC) subsets after uptake of naturally formed antigen-antibody complexes in vivo. We observed that murine splenic DC subsets were able to present antigen in vivo for at least a week. After ex vivo isolation of four APC subsets, the presence of antigen in the storage compartments was visualized by confocal microscopy. Although all APC subsets stored antigen for many days, their ability and kinetics in antigen presentation was remarkably different. CD8α+ DCs showed sustained MHC class I-peptide specific CD8+ T-cell activation for more than 4 days. CD8α- DCs also presented antigenic peptides in MHC class I but presentation decreased after 48 h. In contrast, only the CD8α- DCs were able to present antigen in MHC class II to specific CD4+ T cells. Plasmacytoid DCs and macrophages were unable to activate any of the two T-cell types despite detectable antigen uptake. These results indicate that naturally occurring DC subsets have functional antigen storage capacity for prolonged T-cell activation and have distinct roles in antigen presentation to specific T cells in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming , Dendritic Cells/immunology , Macrophages/immunology , Animals , Antigen Presentation , CD8 Antigens/metabolism , Cells, Cultured , Dendritic Cells/metabolism , Histocompatibility Antigens Class I/metabolism , Histocompatibility Antigens Class II/metabolism , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Microscopy, Confocal , Spleen/immunology
18.
Bioconjug Chem ; 30(4): 1150-1161, 2019 04 17.
Article in English | MEDLINE | ID: mdl-30865430

ABSTRACT

Simultaneous triggering of Toll-like receptors (TLRs) and NOD-like receptors (NLRs) has previously been shown to synergistically activate monocytes, dendritic cells, and macrophages. We applied these properties in a T-cell vaccine setting by conjugating the NOD2-ligand muramyl-dipeptide (MDP) and TLR2-ligand Pam3CSK4 to a synthetic peptide derived from a model antigen. Stimulation of human DCs with the MDP-peptide-Pam3CSK4 conjugate led to a strongly increased secretion of pro-inflammatory and Th1-type cytokines and chemokines. We further show that the conjugated ligands retain their ability to trigger their respective receptors, while even improving NOD2-triggering. Also, activation of murine DCs was enhanced by the dual triggering, ultimately leading to effective induction of vaccine-specific T cells expressing IFNγ, IL-2, and TNFα. Together, these data indicate that the dual MDP-SLP-Pam3CSK4 conjugate constitutes a chemically well-defined vaccine approach that holds promise for the use in the treatment of virus infections and cancer.


Subject(s)
Dendritic Cells/immunology , Nod2 Signaling Adaptor Protein/immunology , Peptides/immunology , Toll-Like Receptor 2/immunology , Vaccines, Conjugate/immunology , Animals , Cytokines/biosynthesis , Dendritic Cells/cytology , HEK293 Cells , Humans , Mice , Mice, Transgenic , Monocytes/immunology , Vaccines, Conjugate/chemistry
19.
Bioorg Med Chem Lett ; 29(11): 1340-1344, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30952595

ABSTRACT

Covalent linking of immunogenic oligopeptides with synthetic Toll-like receptor ligands is a useful approach to develop self-adjuvanting vaccines. In particular, small-molecule based agonists of Toll-like receptor 7 (TLR7) that are derived from 8-oxo-adenine core are potentially promising because these chemically robust TLR7 ligands can be connected to peptide T-cell epitopes via straightforward solid-phase peptide synthesis. In this contribution we present the synthesis of a Boc-protected 9-benzyl-2-alkoxy-8-oxo-adenine building block and its application in the online solid phase synthesis of three peptide conjugates that differ in the position of the TLR7 ligand within the peptide. The conjugates are able to induce dendritic cell maturation and T cell proliferation while the position of the ligand impacts T cell proliferation potency.


Subject(s)
Adenine/pharmacology , Peptides/pharmacology , Toll-Like Receptor 7/agonists , Vaccines, Synthetic/immunology , Adenine/analogs & derivatives , Adenine/chemistry , Dose-Response Relationship, Drug , Humans , Ligands , Molecular Structure , Peptides/chemistry , Structure-Activity Relationship , Toll-Like Receptor 7/immunology , Vaccines, Synthetic/chemistry
20.
J Immunol ; 198(11): 4235-4243, 2017 06 01.
Article in English | MEDLINE | ID: mdl-28432146

ABSTRACT

Dendritic cells (DCs) are specialized in Ag engulfment via a wide variety of uptake receptors on their cell surface. In the present study we investigated Ag uptake and presentation of in vivo-formed Ag-Ab complexes by i.v. injecting mice with Ag-specific Abs followed by the cognate Ag. We show by this natural Ab-mediated Ag targeting system that uptake by splenic APC subsets is severely hampered in mice lacking complement factor C1q (C1qa-/-). Moreover, no detectable Ag cross-presentation by CD8α+ DCs from C1qa-/- mice was found. On the contrary, Ag uptake was not hampered by APCs in FcγRI/II/III/IV-deficient (FcγR quadruple-/-) mice, and the cross-presentation ability of CD8α+ DCs was not affected. In conclusion, we show that C1q rather than FcγRs controls the Ab-mediated Ag uptake and its presentation by spleen APC subsets to T cells.


Subject(s)
Antigen Presentation , Antigen-Antibody Complex/immunology , Complement C1q/immunology , Dendritic Cells/immunology , Adaptive Immunity , Animals , CD8 Antigens/immunology , Complement C1q/deficiency , Complement C1q/genetics , Cross-Priming , Mice , Mice, Inbred C57BL , Receptors, IgG/immunology
SELECTION OF CITATIONS
SEARCH DETAIL