Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 47
Filter
1.
Anal Chem ; 94(22): 7804-7813, 2022 06 07.
Article in English | MEDLINE | ID: mdl-35616489

ABSTRACT

Feature extraction algorithms are an important class of unsupervised methods used to reduce data dimensionality. They have been applied extensively for time-of-flight secondary ion mass spectrometry (ToF-SIMS) imaging─commonly, matrix factorization (MF) techniques such as principal component analysis have been used. A limitation of MF is the assumption of linearity, which is generally not accurate for ToF-SIMS data. Recently, nonlinear autoencoders have been shown to outperform MF techniques for ToF-SIMS image feature extraction. However, another limitation of most feature extraction methods (including autoencoders) that is particularly important for hyperspectral data is that they do not consider spatial information. To address this limitation, we describe the application of the convolutional autoencoder (CNNAE) to hyperspectral ToF-SIMS imaging data. The CNNAE is an artificial neural network developed specifically for hyperspectral data that uses convolutional layers for image encoding, thereby explicitly incorporating pixel neighborhood information. We compared the performance of the CNNAE with other common feature extraction algorithms for two biological ToF-SIMS imaging data sets. We investigated the extracted features and used the dimensionality-reduced data to train additional ML algorithms. By converting two-dimensional convolutional layers to three-dimensional (3D), we also showed how the CNNAE can be extended to 3D ToF-SIMS images. In general, the CNNAE produced features with significantly higher contrast and autocorrelation than other techniques. Furthermore, histologically recognizable features in the data were more accurately represented. The extension of the CNNAE to 3D data also provided an important proof of principle for the analysis of more complex 3D data sets.


Subject(s)
Neural Networks, Computer , Spectrometry, Mass, Secondary Ion , Algorithms , Principal Component Analysis , Spectrometry, Mass, Secondary Ion/methods
2.
Subcell Biochem ; 94: 499-520, 2020.
Article in English | MEDLINE | ID: mdl-32189313

ABSTRACT

C-reactive protein (CRP) is an evolutionary highly conserved member of the pentraxin superfamily of proteins. CRP is widely used as a marker of inflammation, infection and for risk stratification of cardiovascular events. However, there is now a large body of evidence, that continues to evolve, detailing that CRP directly mediates inflammatory reactions and the innate immune response in the context of localised tissue injury. These data support the concept that the pentameric conformation of CRP dissociates into pro-inflammatory CRP isoforms termed pCRP* and monomeric CRP. These pro-inflammatory CRP isoforms undergo conformational changes that facilitate complement binding and immune cell activation and therefore demonstrate the ability to trigger complement activation, activate platelets, monocytes and endothelial cells. The dissociation of pCRP occurs on the surface of necrotic, apoptotic, and ischaemic cells, regular ß-sheet structures such as ß-amyloid, the membranes of activated cells (e.g., platelets, monocytes, and endothelial cells), and/or the surface of microparticles, the latter by binding to phosphocholine. Therefore, the deposition and localisation of these pro-inflammatory isoforms of CRP have been demonstrated to amplify inflammation and tissue damage in a broad range of clinical conditions including ischaemia/reperfusion injury, Alzheimer's disease, age-related macular degeneration and immune thrombocytopaenia. Given the potentially broad relevance of CRP to disease pathology, the development of inhibitors of CRP remains an area of active investigation, which may pave the way for novel therapeutics for a diverse range of inflammatory diseases.


Subject(s)
C-Reactive Protein/chemistry , C-Reactive Protein/metabolism , Conserved Sequence , Evolution, Molecular , Inflammation/metabolism , Inflammation/pathology , Biomarkers/chemistry , Biomarkers/metabolism , Humans , Protein Isoforms/chemistry , Protein Isoforms/metabolism
3.
Haematologica ; 103(4): 655-665, 2018 04.
Article in English | MEDLINE | ID: mdl-29351987

ABSTRACT

Chemotherapy and hematopoietic stem cell transplantation are effective treatments for most Hodgkin lymphoma patients, however there remains a need for better tumor-specific target therapy in Hodgkin lymphoma patients with refractory or relapsed disease. Herein, we demonstrate that membrane CD83 is a diagnostic and therapeutic target, highly expressed in Hodgkin lymphoma cell lines and Hodgkin and Reed-Sternberg cells in 29/35 (82.9%) Hodgkin lymphoma patient lymph node biopsies. CD83 from Hodgkin lymphoma tumor cells was able to trogocytose to surrounding T cells and, interestingly, the trogocytosing CD83+T cells expressed significantly more programmed death-1 compared to CD83-T cells. Hodgkin lymphoma tumor cells secreted soluble CD83 that inhibited T-cell proliferation, and anti-CD83 antibody partially reversed the inhibitory effect. High levels of soluble CD83 were detected in Hodgkin lymphoma patient sera, which returned to normal in patients who had good clinical responses to chemotherapy confirmed by positron emission tomography scans. We generated a human anti-human CD83 antibody, 3C12C, and its toxin monomethyl auristatin E conjugate, that killed CD83 positive Hodgkin lymphoma cells but not CD83 negative cells. The 3C12C antibody was tested in dose escalation studies in non-human primates. No toxicity was observed, but there was evidence of CD83 positive target cell depletion. These data establish CD83 as a potential biomarker and therapeutic target in Hodgkin lymphoma.


Subject(s)
Antigens, CD/blood , Biomarkers, Tumor/blood , Hodgkin Disease/drug therapy , Immunoglobulins/blood , Membrane Glycoproteins/blood , Molecular Targeted Therapy/methods , Adolescent , Adult , Aged , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Antigens, CD/immunology , Female , Hodgkin Disease/diagnosis , Humans , Immunoglobulins/immunology , Male , Membrane Glycoproteins/immunology , Middle Aged , Salvage Therapy/methods , T-Lymphocytes/cytology , Young Adult , CD83 Antigen
4.
Molecules ; 23(9)2018 Sep 02.
Article in English | MEDLINE | ID: mdl-30200528

ABSTRACT

Peptide-based vaccines for cancer have many advantages however, for optimization these immunogens should incorporate peptide epitopes that induce CD8, as well as CD4 responses, antibody and long term immunity. Cell penetrating peptides (CPP) with a capacity of cytosolic delivery have been used to deliver antigenic peptides and proteins to antigen presenting cells to induce cytotoxic T cell, helper T cell and humoral responses in mice. For this study, a tripartite CPP including a mucin 1 (MUC1) variable number of tandem repeat (VNTR) containing multiple T cell epitopes and tetanus toxoid universal T helper epitope peptide (tetCD4) was synthesised (AntpMAPMUC1tet) and immune responses investigated in mice. Mice vaccinated with AntpMAPMUC1tet + CpG show enhanced antigen-specific interferon-gamma (IFN-γ) and IL-4 T cell responses compared with AntpMAPMUC1tet vaccination alone and induced a Th1 response, characterised by a higher ratio of IgG2a antibody/IgG1 antibodies. Furthermore, vaccination generated long term MUC1-specific antibody and T cell responses and delayed growth of MUC1+ve tumours in mice. This data demonstrates the efficient delivery of branched multiple antigen peptides incorporating CPP and that the addition of CpG augments immune responses.


Subject(s)
Cell-Penetrating Peptides/immunology , Epitopes, T-Lymphocyte/immunology , Minisatellite Repeats/genetics , Mucin-1/genetics , T-Lymphocytes, Helper-Inducer/immunology , Amino Acid Sequence , Animals , Antibody Specificity/drug effects , Antigen-Presenting Cells/metabolism , Bone Marrow Cells/drug effects , Bone Marrow Cells/metabolism , Cell Differentiation , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/pharmacology , Cytotoxicity, Immunologic/drug effects , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Endocytosis , Epitopes , Epitopes, T-Lymphocyte/chemistry , Female , HLA-A2 Antigen/metabolism , Immunity, Cellular/drug effects , Immunization , Interferon-gamma/metabolism , Interleukin-4/metabolism , Mice, Inbred C57BL , Mice, Transgenic , Neoplasms/immunology , Neoplasms/pathology , Oligodeoxyribonucleotides/metabolism , Tetanus Toxin/chemistry
5.
J Immunol ; 192(2): 792-803, 2014 Jan 15.
Article in English | MEDLINE | ID: mdl-24342805

ABSTRACT

Little is known of the impact of Fc receptor (FcR) polymorphism in macaques on the binding of human (hu)IgG, and nothing is known of this interaction in the pig-tailed macaque (Macaca nemestrina), which is used in preclinical evaluation of vaccines and therapeutic Abs. We defined the sequence and huIgG binding characteristics of the M. nemestrina activating FcγRIIa (mnFcγRIIa) and inhibitory FcγRIIb (mnFcγRIIb) and predicted their structures using the huIgGFc/huFcγRIIa crystal structure. Large differences were observed in the binding of huIgG by mnFcγRIIa and mnFcγRIIb compared with their human FcR counterparts. MnFcγRIIa has markedly impaired binding of huIgG1 and huIgG2 immune complexes compared with huFcγRIIa (His(131)). In contrast, mnFcγRIIb has enhanced binding of huIgG1 and broader specificity, as, unlike huFcγRIIb, it avidly binds IgG2. Mutagenesis and molecular modeling of mnFcγRIIa showed that Pro(159) and Tyr(160) impair the critical FG loop interaction with huIgG. The enhanced binding of huIgG1 and huIgG2 by mnFcγRIIb was shown to be dependent on His(131) and Met(132). Significantly, both His(131) and Met(132) are conserved across FcγRIIb of rhesus and cynomolgus macaques. We identified functionally significant polymorphism of mnFcγRIIa wherein proline at position 131, also an important polymorphic site in huFcγRIIa, almost abolished binding of huIgG2 and huIgG1 and reduced binding of huIgG3 compared with mnFcγRIIa His(131). These marked interspecies differences in IgG binding between human and macaque FcRs and polymorphisms within species have implications for preclinical evaluation of Abs and vaccines in macaques.


Subject(s)
Immunoglobulin G/metabolism , Macaca nemestrina/genetics , Macaca nemestrina/metabolism , Polymorphism, Genetic/genetics , Receptors, IgG/genetics , Receptors, IgG/metabolism , Amino Acid Sequence , Animals , Humans , Molecular Sequence Data , Protein Binding/genetics , Sequence Alignment
6.
Molecules ; 20(8): 14033-50, 2015 Aug 03.
Article in English | MEDLINE | ID: mdl-26247926

ABSTRACT

Cell penetrating peptides (CPP), including the TAT peptide from the human immunodeficiency virus transactivator of transcription (HIV-TAT) protein and penetratin from Drosophila Antennapedia homeodomain protein, translocate various cargos including peptides and proteins across cellular barriers. This mode of delivery has been harnessed by our group and others to deliver antigenic proteins or peptides into the cytoplasm of antigen processing cells (APC) such as monocyte-derived dendritic cells (MoDC). Antigens or T cell epitopes delivered by CPP into APC in vivo generate antigen-specific cytotoxic T cell and helper T cell responses in mice. Furthermore, mice immunised with these peptides or proteins are protected from a tumour challenge. The functional properties of CPP are dependent on the various cargos being delivered and the target cell type. Despite several studies demonstrating superior immunogenicity of TAT and Antp-based immunogens, none has compared the immunogenicity of antigens delivered by TAT and Antp CPP. In the current study we demonstrate that a cytotoxic T cell epitope from the mucin 1 (MUC1) tumour associated antigen, when delivered by TAT or Antp, generates identical immune responses in mice resulting in specific MUC1 T cell responses as measured by in vivo CTL assays, IFNγ ELISpot assays and prophylactic tumour protection.


Subject(s)
Carrier Proteins/pharmacology , Cell-Penetrating Peptides/pharmacology , Cytotoxicity, Immunologic/drug effects , Epitopes, T-Lymphocyte/immunology , T-Lymphocytes, Cytotoxic/drug effects , tat Gene Products, Human Immunodeficiency Virus/pharmacology , Amino Acid Sequence , Animals , Bone Marrow Cells/cytology , Carrier Proteins/chemistry , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Endocytosis/drug effects , Immunity, Cellular/drug effects , Immunization , Interferon-gamma/metabolism , Lymphocyte Activation/drug effects , Mice, Inbred C57BL , Molecular Sequence Data , Mucin-1/metabolism , Neoplasms/pathology , Ovalbumin/immunology , tat Gene Products, Human Immunodeficiency Virus/chemistry
7.
Eur J Immunol ; 43(5): 1208-19, 2013 May.
Article in English | MEDLINE | ID: mdl-23420539

ABSTRACT

Previous studies on the role of the tetraspanin CD37 in cellular immunity appear contradictory. In vitro approaches indicate a negative regulatory role, whereas in vivo studies suggest that CD37 is necessary for optimal cellular responses. To resolve this discrepancy, we studied the adaptive cellular immune responses of CD37(-/-) mice to intradermal challenge with either tumors or model antigens and found that CD37 is essential for optimal cell-mediated immunity. We provide evidence that an increased susceptibility to tumors observed in CD37(-/-) mice coincides with a striking failure to induce antigen-specific IFN-γ-secreting T cells. We also show that CD37 ablation impairs several aspects of DC function including: in vivo migration from skin to draining lymph nodes; chemo-tactic migration; integrin-mediated adhesion under flow; the ability to spread and form actin protrusions and in vivo priming of adoptively transferred naïve T cells. In addition, multiphoton microscopy-based assessment of dermal DC migration demonstrated a reduced rate of migration and increased randomness of DC migration in CD37(-/-) mice. Together, these studies are consistent with a model in which the cellular defect that underlies poor cellular immune induction in CD37(-/-) mice is impaired DC migration.


Subject(s)
Antigens, CD/immunology , Antigens, Neoplasm/immunology , Cell Movement/immunology , Dendritic Cells/immunology , Immunity, Cellular , Tetraspanins/immunology , Adaptive Immunity , Adoptive Transfer , Animals , Antigens, CD/genetics , Antigens, Neoplasm/genetics , Cell Adhesion/immunology , Cell Proliferation , Dendritic Cells/pathology , Female , Gene Expression , Interferon-gamma/biosynthesis , Interferon-gamma/immunology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphoma, T-Cell/genetics , Lymphoma, T-Cell/immunology , Lymphoma, T-Cell/pathology , Mice , Mice, Knockout , Microscopy, Confocal , Neoplasm Transplantation , Skin/immunology , Skin/pathology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/transplantation , Tetraspanins/deficiency , Tetraspanins/genetics
8.
Theranostics ; 14(8): 3267-3281, 2024.
Article in English | MEDLINE | ID: mdl-38855181

ABSTRACT

Background: Myocardial infarction (MI) as a consequence of atherosclerosis-associated acute thrombosis is a leading cause of death and disability globally. Antiplatelet and anticoagulant drugs are standard therapies in preventing and treating MI. However, all clinically used drugs are associated with bleeding complications, which ultimately limits their use in patients with a high risk of bleeding. We have developed a new recombinant drug, targ-HSA-TAP, that combines targeting and specific inhibition of activated platelets as well as anticoagulation. This drug is designed and tested for a prolonged circulating half-life, enabling unique thromboprophylaxis without bleeding complications. Methods: Targ-HSA-TAP combines a single-chain antibody (scFv) that targets activated glycoprotein IIb/IIIa on activated platelets, human serum albumin (HSA) for prolonged circulation, and tick anticoagulant peptide (TAP) for coagulation FX inhibition. A non-binding scFv is employed as a non-targeting control (non-targ-HSA-TAP). Its efficacy was investigated in vivo using murine models of acute thrombosis and cardiac ischemia-reperfusion (I/R) injury. Results: Our experiments confirmed the targeting specificity of targ-HSA-TAP to activated platelets and demonstrated effective prevention of platelet aggregation and thrombus formation, as well as FXa inhibition in vitro. Thromboprophylactic administration of targ-HSA-TAP subcutaneously in mice prevented occlusion of the carotid artery after ferric chloride injury as compared to non-targ-HSA-TAP and PBS-control treated mice. By comparing the therapeutic outcomes between targ-TAP and targ-HSA-TAP, we demonstrate the significant improvements brought by the HSA fusion in extending the drug's half-life and enhancing its therapeutic window for up to 16 h post-administration. Importantly, tail bleeding time was not prolonged with targ-HSA-TAP in contrast to the clinically used anticoagulant enoxaparin. Furthermore, in a murine model of cardiac I/R injury, mice administered targ-HSA-TAP 10 h before injury demonstrated preserved cardiac function, with significantly higher ejection fraction and fractional shortening, as compared to the non-targ-HSA-TAP and PBS control groups. Advanced strain analysis revealed reduced myocardial deformation and histology confirmed a reduced infarct size in targ-HSA-TAP treated mice compared to control groups. Conclusion: The inclusion of HSA represents a significant advancement in the design of targeted therapeutic agents for thromboprophylaxis. Our activated platelet-targeted targ-HSA-TAP is a highly effective antithrombotic drug with both anticoagulant and antiplatelet effects while retaining normal hemostasis. The long half-life of targ-HSA-TAP provides the unique opportunity to use this antithrombotic drug for more effective, long-lasting and safer anti-thrombotic prophylaxis. In cases where MI occurs, this prophylactic strategy reduces thrombus burden and effectively reduces cardiac I/R injury.


Subject(s)
Blood Platelets , Hemorrhage , Serum Albumin, Human , Thrombosis , Animals , Mice , Thrombosis/prevention & control , Thrombosis/drug therapy , Humans , Hemorrhage/prevention & control , Blood Platelets/drug effects , Blood Platelets/metabolism , Disease Models, Animal , Male , Anticoagulants/pharmacology , Anticoagulants/therapeutic use , Single-Chain Antibodies/pharmacology , Single-Chain Antibodies/therapeutic use , Myocardial Reperfusion Injury/prevention & control , Myocardial Reperfusion Injury/drug therapy , Myocardial Infarction/drug therapy , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacology , Recombinant Fusion Proteins/therapeutic use
9.
J Immunol ; 184(6): 2863-72, 2010 Mar 15.
Article in English | MEDLINE | ID: mdl-20176741

ABSTRACT

Reactive oxygen species (ROS) have been implicated in various physiological activities. However, their role in dendritic cell (DC) activation and generation has not been investigated. Using the bone marrow-derived GM-CSF-induced ex vivo DC model, we characterize how induction of ROS correlates with inflammatory DC functionality and expansion. We describe that the functionality of GM-CSF-induced DCs is distinct in two developmental stages. Whereas division of DC-committed hematopoietic progenitor cells (HPCs) neared completion by day 6, the level of ROS soared after day 4. Day 3 ROS(lo) DCs were highly responsive to TLR stimuli such as LPS and zymosan by rapid upregulation of CD80, CD86, and MHC class II, in contrast to the low response of day 6 ROS(hi) DCs. ROS(hi) DCs could not initiate and sustain a significant level of NF-kappaB phosphorylation in response to LPS and zymosan, although demonstrating hyperactivation of p38 MAPK by LPS, in a fashion disparate to ROS(lo) DCs. ROS(lo) DCs stimulated a higher level of allogeneic and OVA-specific T cell proliferative responses, although ROS(hi) DCs were much more proficient in processing OVA. In response to pathogenic stimuli, ROS(hi) DCs also demonstrated rapid cellular adhesion and H(2)O(2) release, indicating their role in immediate microbial targeting. Moreover, HPC expansion and DC generation were dependent on the surge of ROS in an NADPH oxidase-independent manner. These findings point to the potential role of cellular ROS in mediating functionality and development of DCs from HPCs during inflammation.


Subject(s)
Bone Marrow Cells/metabolism , Bone Marrow Cells/pathology , Cell Differentiation/immunology , Dendritic Cells/metabolism , Dendritic Cells/pathology , Inflammation Mediators/physiology , Reactive Oxygen Species/metabolism , Amino Acid Sequence , Animals , Bone Marrow Cells/immunology , Cells, Cultured , Dendritic Cells/immunology , Hematopoietic Stem Cells/immunology , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/pathology , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Transgenic , Molecular Sequence Data , Up-Regulation/immunology
10.
Biochim Biophys Acta ; 1798(12): 2286-95, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20478265

ABSTRACT

Cell penetrating peptides (CPP) represent a novel approach to facilitate cytoplasmic delivery of macromolecules. The DNA binding domain of Drosophila Antennapedia contains 60 amino acids and consists of 3 α-helices, with internalizing activity mapped to a 16-amino acid peptide penetratin (Antp) within the third α-helix. Here, we report on the use of penetratin to deliver a multiple antigen peptide (MAP) incorporating the immunodominant CD8 epitope of ovalbumin, SIINFEKL (MAPOVACD8). We demonstrate that penetratin linked to the MAPOVACD8 construct either by a disulfide (SS) or thioether (SC) linkage promotes the uptake, cross presentation and subsequent in vivo proliferation and generation of OVACD8 (SIINFEKL)-specific T cells. The MAPOVACD8 construct without penetratin is not presented by MHC class I molecules nor does it generate an in vivo IFN-γ response in C57BL/6 mice. Moreover, we clearly define the uptake and intracellular processing pathways of AntpMAPOVACD8 SS and SC revealing the majority of AntpMAPOVACD8 is taken up by DC via an endocytic, proteasome and tapasin independent mechanism. We also show that the uptake mechanism of AntpMAPOVACD8 is dose dependent and uptake or intracellular processing is not altered by the type of chemical linkage.


Subject(s)
Carrier Proteins , Cell-Penetrating Peptides , Epitopes , Ovalbumin , T-Lymphocytes/immunology , Animals , Antigen Presentation/drug effects , Antigen Presentation/immunology , Carrier Proteins/chemistry , Carrier Proteins/immunology , Carrier Proteins/pharmacology , Cell Proliferation/drug effects , Cell-Penetrating Peptides/chemistry , Cell-Penetrating Peptides/immunology , Cell-Penetrating Peptides/pharmacology , Dendritic Cells/immunology , Dose-Response Relationship, Drug , Dose-Response Relationship, Immunologic , Drosophila , Endocytosis/drug effects , Endocytosis/immunology , Epitopes/chemistry , Epitopes/immunology , Epitopes/pharmacology , Histocompatibility Antigens Class I/immunology , Interferon-gamma/immunology , Membrane Transport Proteins/immunology , Mice , Ovalbumin/chemistry , Ovalbumin/immunology , Ovalbumin/pharmacology , Proteasome Endopeptidase Complex/immunology , Protein Structure, Secondary
11.
Biochim Biophys Acta ; 1805(1): 25-34, 2010 Jan.
Article in English | MEDLINE | ID: mdl-19782720

ABSTRACT

Recent years have seen a surge in interest in cell-penetrating peptides (CPP) as an efficient means for delivering therapeutic targets into cellular compartments. The cell membrane is impermeable to hydrophilic substances yet linking to CPP can facilitate delivery into cells. Thus the unique translocatory property of CPP ensures they remain an attractive carrier, with the capacity to deliver cargoes in an efficient manner having applications in drug delivery, gene transfer and DNA vaccination. Fundamental for an effective vaccine is the delivery of antigen epitopes to antigen-presenting cells, ensuing processing and presentation and induction of an immune response. Vaccination with proteins or synthetic peptides incorporating CTL epitopes have proven limited due to the failure for exogenous antigens to be presented efficiently to T cells. Linking of antigens to CPP overcomes such obstacles by facilitating cellular uptake, processing and presentation of exogenous antigen for the induction of potent immune responses. This review will encompass the various strategies for the delivery of whole proteins, T cell epitopes and preclinical studies utilizing CPP for cancer vaccines.


Subject(s)
Cancer Vaccines/administration & dosage , Carrier Proteins/administration & dosage , Drug Delivery Systems , Neoplasms/drug therapy , Peptide Fragments/administration & dosage , Cell-Penetrating Peptides , Humans , Neoplasms/immunology
12.
Immunol Cell Biol ; 89(8): 904-13, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21383765

ABSTRACT

Cytoplasmic delivery and cross-presentation of proteins and peptides is necessary for processing and presentation of antigens for the generation of cytotoxic T cells. We previously described the use of the 16 amino acid peptide penetratin from the Drosophila Antennapedia homeodomain (penetratin, Antp) to transport cytotoxic T lymphocyte epitopes derived from ovalbumin (OVA) or the Mucin-1 tumor-associated antigen into cells. We have now shown that penetratin covalently conjugated to OVA protein and linked in tandem to CD4(+) and/or CD8(+) T-cell epitopes from OVA-stimulated T cells in vitro (B3Z T-cell hybridoma and OT-I and OT-II T cells). The induction of these responses was directly mediated by the penetratin peptide as linking a nonspecific 16-mer peptide to OVA or mixing did not induce CD8(+) or CD4(+) T-cell responses in vitro. Furthermore, interferon (IFN)-γ-secreting CD4(+) and CD8(+) T cells were induced which suppressed B16.OVA tumor growth in C57BL/6 mice. Tumor protection was mediated by a CD8(+) T-cell-dependent mechanism and did not require CD4(+) help to protect mice 7 days after a boost immunization. Alternatively, 40 days after a boost immunization, the presence of CD4(+) help enhanced antigen-specific IFN-γ-secreting CD8(+) T cells and tumor protection in mice challenged with B16.OVA. Long-term CD8 responses were equally enhanced by antigen-specific and universal CD4 help. In addition, immunization with AntpOVA significantly delayed growth of B16.OVA tumors in mice in a tumor therapy model.


Subject(s)
Antigen Presentation , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/immunology , Neoplasms/immunology , Animals , Antennapedia Homeodomain Protein/immunology , Antennapedia Homeodomain Protein/metabolism , Antigens, Neoplasm/immunology , CD4 Antigens/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Carrier Proteins , Cell-Penetrating Peptides , Drosophila , Drosophila Proteins/immunology , Interferon-gamma/biosynthesis , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , Mucin-1/immunology , Mucin-1/metabolism , Ovalbumin/immunology , Ovalbumin/metabolism , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Helper-Inducer/immunology
13.
Front Immunol ; 12: 666813, 2021.
Article in English | MEDLINE | ID: mdl-34759915

ABSTRACT

FcγR activity underpins the role of antibodies in both protective immunity and auto-immunity and importantly, the therapeutic activity of many monoclonal antibody therapies. Some monoclonal anti-FcγR antibodies activate their receptors, but the properties required for cell activation are not well defined. Here we examined activation of the most widely expressed human FcγR; FcγRIIa, by two non-blocking, mAbs, 8.26 and 8.2. Crosslinking of FcγRIIa by the mAb F(ab')2 regions alone was insufficient for activation, indicating activation also required receptor engagement by the Fc region. Similarly, when mutant receptors were inactivated in the Fc binding site, so that intact mAb was only able to engage receptors via its two Fab regions, again activation did not occur. Mutation of FcγRIIa in the epitope recognized by the agonist mAbs, completely abrogated the activity of mAb 8.26, but mAb 8.2 activity was only partially inhibited indicating differences in receptor recognition by these mAbs. FcγRIIa inactivated in the Fc binding site was next co-expressed with the FcγRIIa mutated in the epitope recognized by the Fab so that each mAb 8.26 molecule can contribute only three interactions, each with separate receptors, one via the Fc and two via the Fab regions. When the Fab and Fc binding were thus segregated onto different receptor molecules receptor activation by intact mAb did not occur. Thus, receptor activation requires mAb 8.26 Fab and Fc interaction simultaneously with the same receptor molecules. Establishing the molecular nature of FcγR engagement required for cell activation may inform the optimal design of therapeutic mAbs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antibody-Dependent Cell Cytotoxicity/immunology , Immunoglobulin Fc Fragments/metabolism , Receptors, IgG/agonists , Receptors, IgG/metabolism , Binding Sites , Epitopes/genetics , Epitopes/immunology , Humans , Immunoglobulin Fab Fragments/immunology , Immunoglobulin Fc Fragments/immunology , Mutation , Phosphorylation , Platelet Activation , Protein Binding , Receptors, Fc , Receptors, IgG/genetics
14.
J Immunol ; 181(4): 2455-64, 2008 Aug 15.
Article in English | MEDLINE | ID: mdl-18684936

ABSTRACT

The evidence that dendritic cell (DC) subsets produce differential cytokines in response to specific TLR stimulation is robust. However, the role of TLR stimulation in Ag presentation and phenotypic maturation among DC subsets is not clear. Through the adjuvanticity of a novel mannosylated Ag, mannosylated dendrimer OVA (MDO), as a pathogen-associated molecular pattern Ag, we characterized the functionality of GM-CSF/IL-4-cultured bone marrow DC and Flt3 ligand (Flt3-L) DC subsets by Ag presentation and maturation assays. It was demonstrated that both bone marrow DCs and Flt3-L DCs bound, processed, and presented MDO effectively. However, while Flt3-L CD24(high) (conventional CD8(+) equivalent) and CD11b(high) (CD8(-) equivalent) DCs were adept at MDO processing by MHC class I and II pathways, respectively, CD45RA(+) plasmacytoid DCs presented MDO poorly to T cells. Successful MDO presentation was largely dependent on competent TLR4 for Ag localization and morphological/phenotypic maturation of DC subsets, despite the indirect interaction of MDO with TLR4. Furthermore, Toll/IL-1 receptor-domain-containing adaptor-inducing IFN-beta, but not MyD88, as a TLR4 signaling modulator was indispensable for MDO-induced DC maturation and Ag presentation. Taken together, our findings suggest that DC subsets differentially respond to a pathogen-associated molecular pattern-associated Ag depending on the intrinsic programming and TLRs expressed. Optimal functionality of DC subsets in Ag presentation necessitates concomitant TLR signaling critical for efficient Ag localization and processing.


Subject(s)
Adjuvants, Immunologic/physiology , Antigens/metabolism , Cell Differentiation/immunology , Dendritic Cells/cytology , Dendritic Cells/immunology , Mannose/metabolism , Ovalbumin/immunology , Toll-Like Receptor 4/physiology , Amino Acid Sequence , Animals , Antigen Presentation/immunology , Antigens/immunology , Bone Marrow Cells/classification , Bone Marrow Cells/immunology , Bone Marrow Cells/metabolism , Cell Communication/genetics , Cell Communication/immunology , Cell Differentiation/genetics , Cells, Cultured , Dendritic Cells/classification , Dendritic Cells/metabolism , Immunophenotyping , Mice , Mice, Inbred C3H , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Molecular Sequence Data , Ovalbumin/chemical synthesis , Ovalbumin/metabolism , Toll-Like Receptor 4/deficiency , Toll-Like Receptor 4/genetics
15.
Blood Adv ; 4(7): 1206-1216, 2020 04 14.
Article in English | MEDLINE | ID: mdl-32215656

ABSTRACT

Allogeneic hematopoietic stem cell transplantation (allo-HSCT) significantly reduces the rate of relapse in acute myeloid leukemia (AML) but comes at the cost of significant treatment-related mortality. Despite the reduction in relapse overall, it remains common, especially in high-risk groups. The outcomes for patients who relapse after transplant remains very poor. A large proportion of the morbidity that prevents most patients from accessing allo-HSCT is due to toxic nonspecific conditioning agents that are required to remove recipient hematopoietic stem and progenitor cells (HSPCs), allowing for successful donor engraftment. CD300f is expressed evenly across HSPC subtypes. CD300f has transcription and protein expression equivalent to CD33 on AML. We have developed an anti-CD300f antibody that efficiently internalizes into target cells. We have generated a highly potent anti-CD300f antibody-drug conjugate (ADC) with a pyrrolobenzodiazepine warhead that selectively depletes AML cell lines and colony forming units in vitro. The ADC synergizes with fludarabine, making it a natural combination to use in a minimal toxicity conditioning regimen. Our ADC prolongs the survival of mice engrafted with human cell lines and depletes primary human AML engrafted with a single injection. In a humanized mouse model, a single injection of the ADC depletes CD34+ HSPCs and CD34+CD38-CD90+ hematopoietic stem cells. This work establishes an anti-CD300f ADC as an attractive potential therapeutic that, if validated in transplant models using a larger cohort of primary AML samples, will reduce relapse rate and toxicity for patients with AML undergoing allo-HSCT.


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukemia, Myeloid, Acute , Animals , Humans , Leukemia, Myeloid, Acute/therapy , Mice , Retrospective Studies , Transplantation Conditioning , Transplantation, Homologous
16.
Mol Immunol ; 45(2): 307-19, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17673295

ABSTRACT

A disulphide-constrained peptide that binds to the low affinity Fc receptor, FcgammaRIIa (CD32) has been identified and its structure solved by NMR. Linear (7-mer and 12-mer) and disulphide-constrained (7-mer) phage display peptide libraries were panned on recombinant soluble FcgammaRIIa genetically fused to HSA (HSA-FcgammaRIIa). Peptides were isolated only from the constrained peptide library and these contained the consensus sequence, CWPGWxxC. Phage clones displaying variants of the peptide consensus sequence bound to FcgammaRIIa and the strongest binding clone C7C1 (CWPGWDLNC) competed with IgG for binding to FcgammaRIIa and was inhibited from binding to FcgammaRIIa by the FcgammaRIIa-blocking antibody, IV.3, suggesting that C7C1 and IgG share related binding sites on FcgammaRIIa. A synthetic disulphide-constrained peptide, pep-C7C1 bound to FcgammaRIIa by biosensor analysis, albeit with low affinity (KD approximately 100microM). It was significant that the FcgammaRIIa consensus peptide sequence contained a Proline (Pro3), which when substituted with alanine abrogated FcgammaRIIa binding, consistent with Pro3 contributing to receptor binding. Upon binding of IgG and IgE to their respective Fc receptors (FcgammaRs and FcepsilonRI) Pro329 in the Fc makes a critical interaction with two highly conserved Trp residues (Trp90 and Trp113) of the FcRs. The NMR structure of pep-C7C1 revealed a stabilizing type II beta-turn between Trp2 and Trp5, with Pro3 solvent exposed. Modelling of the pep-C7C1 structure in complex with FcgammaRIIa suggests that Pro3 of C7C1 binds to FcgammaRIIa by inserting between Trp90 and Trp113 of FcgammaRIIa thereby mimicking the molecular interaction made between FcgammaRIIa and IgG.


Subject(s)
Antigens, CD/metabolism , Immunoglobulin G/metabolism , Molecular Mimicry , Peptides/metabolism , Receptors, IgG/metabolism , Amino Acid Sequence , Antigens, CD/chemistry , Bacteriophages , Biosensing Techniques , Clone Cells , Consensus Sequence , Disulfides/metabolism , Enzyme-Linked Immunosorbent Assay , Humans , Magnetic Resonance Spectroscopy , Models, Molecular , Molecular Sequence Data , Mutagenesis , Peptide Library , Peptides/chemistry , Protein Binding , Protein Structure, Secondary , Receptors, IgE/metabolism , Receptors, IgG/chemistry , Serum Albumin/chemistry , Serum Albumin/metabolism , Surface Properties
17.
Theranostics ; 9(4): 1154-1169, 2019.
Article in English | MEDLINE | ID: mdl-30867822

ABSTRACT

Rationale: Platelets are increasingly recognized as mediators of tumor growth and metastasis. Hypothesizing that activated platelets in the tumor microenvironment provide a targeting epitope for tumor-directed chemotherapy, we developed an antibody-drug conjugate (ADC), comprised of a single-chain antibody (scFv) against the platelet integrin GPIIb/IIIa (scFvGPIIb/IIIa) linked to the potent chemotherapeutic microtubule inhibitor, monomethyl auristatin E (MMAE). Methods: We developed an ADC comprised of three components: 1) A scFv which specifically binds to the high affinity, activated integrin GPIIb/IIIa on activated platelets. 2) A highly potent microtubule inhibitor, monomethyl auristatin E. 3) A drug activation/release mechanism using a linker cleavable by cathepsin B, which we demonstrate to be abundant in the tumor microenvironment. The scFvGPIIb/IIIa-MMAE was first conjugated with Cyanine7 for in vivo imaging. The therapeutic efficacy of the scFvGPIIb/IIIa-MMAE was then tested in a mouse metastasis model of triple negative breast cancer. Results: In vitro studies confirmed that this ADC specifically binds to activated GPIIb/IIIa, and cathepsin B-mediated drug release/activation resulted in tumor cytotoxicity. In vivo fluorescence imaging demonstrated that the newly generated ADC localized to primary tumors and metastases in a mouse xenograft model of triple negative breast cancer, a difficult to treat tumor for which a selective tumor-targeting therapy remains to be clinically established. Importantly, we demonstrated that the scFvGPIIb/IIIa-MMAE displays marked efficacy as an anti-cancer agent, reducing tumor growth and preventing metastatic disease, without any discernible toxic effects. Conclusion: Here, we demonstrate the utility of a novel ADC that targets a potent cytotoxic drug to activated platelets and specifically releases the cytotoxic agent within the confines of the tumor. This unique targeting mechanism, specific to the tumor microenvironment, holds promise as a novel therapeutic approach for the treatment of a broad range of primary tumors and metastatic disease, particularly for tumors that lack specific molecular epitopes for drug targeting.


Subject(s)
Antineoplastic Agents/administration & dosage , Blood Platelets/metabolism , Immunoconjugates/administration & dosage , Molecular Targeted Therapy/methods , Oligopeptides/administration & dosage , Triple Negative Breast Neoplasms/drug therapy , Tumor Microenvironment , Animals , Antineoplastic Agents/metabolism , Disease Models, Animal , Immunoconjugates/metabolism , Mice , Neoplasm Metastasis/drug therapy , Neoplasm Transplantation , Oligopeptides/metabolism , Platelet Glycoprotein GPIIb-IIIa Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/immunology , Single-Chain Antibodies/immunology , Transplantation, Heterologous , Treatment Outcome
18.
Clin Cancer Res ; 12(3 Pt 1): 869-77, 2006 Feb 01.
Article in English | MEDLINE | ID: mdl-16467101

ABSTRACT

PURPOSE: Tumor antigen-loaded dendritic cells show promise for cancer immunotherapy. This phase I study evaluated immunization with autologous dendritic cells pulsed with mannan-MUC1 fusion protein (MFP) to treat patients with advanced malignancy. EXPERIMENTAL DESIGN: Eligible patients had adenocarcinoma expressing MUC1, were of performance status 0 to 1, with no autoimmune disease. Patients underwent leukapheresis to generate dendritic cells by culture ex vivo with granulocyte macrophage colony-stimulating factor and interleukin 4 for 5 days. Dendritic cells were then pulsed overnight with MFP and harvested for reinjection. Patients underwent three cycles of leukapheresis and reinjection at monthly intervals. Patients with clinical benefit were able to continue with dendritic cell-MFP immunotherapy. RESULTS: Ten patients with a range of tumor types were enrolled, with median age of 60 years (range, 33-70 years); eight patients were of performance status 0 and two of performance status 1. Dendritic cell-MFP therapy led to strong T-cell IFNgamma Elispot responses to the vaccine and delayed-type hypersensitivity responses at injection sites in nine patients who completed treatments. Immune responses were sustained at 1 year in monitored patients. Antibody responses were seen in three patients only and were of low titer. Side effects were grade 1 only. Two patients with clearly progressive disease (ovarian and renal carcinoma) at entry were stable after initial therapy and went on to further leukapheresis and dendritic cell-MFP immunotherapy. These two patients have now each completed over 3 years of treatment. CONCLUSIONS: Immunization produced T-cell responses in all patients with evidence of tumor stabilization in 2 of the 10 advanced cancer patients treated. These data support further clinical evaluation of this dendritic cell-MFP immunotherapy.


Subject(s)
Adenocarcinoma/therapy , Cancer Vaccines/administration & dosage , Dendritic Cells/immunology , Immunotherapy , Mannans/administration & dosage , Mucins/administration & dosage , Adenocarcinoma/immunology , Adult , Aged , Antigens, Neoplasm , Cancer Vaccines/immunology , Cancer Vaccines/toxicity , Dendritic Cells/transplantation , Dose-Response Relationship, Immunologic , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunization Schedule , Interferon-gamma/immunology , Interferon-gamma/metabolism , Leukapheresis , Male , Mannans/immunology , Mannans/toxicity , Middle Aged , Mucin-1 , Mucins/immunology , Phenotype , Recombinant Fusion Proteins/administration & dosage , Recombinant Fusion Proteins/immunology , T-Lymphocytes/immunology , Treatment Outcome
19.
Nanomedicine (Lond) ; 12(15): 1873-1889, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28703636

ABSTRACT

The unique chemical and functional properties of nanoparticles can be harnessed for the delivery of large quantities of various therapeutic biomolecules. Active targeting of nanoparticles by conjugating ligands that bind to target cells strongly facilitates accumulation, internalization into target cells and longer retention at the target site, with consequent enhanced therapeutic effects. Recombinant antibodies with high selectivity and availability for a vast range of targets will dominate the future. In this review, we systematically outline the tremendous progress in the conjugation of antibodies to nanoparticles and the clear advantages that recombinant antibodies offer in the therapeutic targeting of nanoparticles. The demonstrated flexibility of recombinant antibody coupling to nanoparticles highlights the bright future of this technology for modern therapeutic nanomedicine.


Subject(s)
Antibodies/chemistry , Antineoplastic Agents/chemistry , Nanoparticles/chemistry , Neoplasms/drug therapy , Recombinant Proteins/chemistry , Animals , Antibodies/pharmacology , Antineoplastic Agents/pharmacology , Drug Carriers , Drug Liberation , Humans , Ligands , Nanomedicine , Recombinant Proteins/pharmacology , Single-Chain Antibodies/chemistry , Single-Chain Antibodies/pharmacology
20.
Theranostics ; 7(10): 2565-2574, 2017.
Article in English | MEDLINE | ID: mdl-28819447

ABSTRACT

Rationale The early detection of primary tumours and metastatic disease is vital for successful therapy and is contingent upon highly specific molecular markers and sensitive, non-invasive imaging techniques. We hypothesized that the accumulation of activated platelets within tumours is a general phenomenon and thus represents a novel means for the molecular imaging of cancer. Here we investigate a unique single chain antibody (scFv), which specifically targets activated platelets, as a novel biotechnological tool for molecular imaging of cancer. Methods The scFvGPIIb/IIIa, which binds specifically to the activated form of the platelet integrin receptor GPIIb/IIIa present on activated platelets, was conjugated to either Cy7, 64Cu or ultrasound-enhancing microbubbles. Using the Cy7 labelled scFvGPIIb/IIIa, fluorescence imaging was performed in mice bearing four different human tumour xenograft models; SKBr3, MDA-MB-231, Ramos and HT-1080 cells. Molecular imaging via PET and ultrasound was performed using the scFvGPIIb/IIIa-64Cu and scFvGPIIb/IIIa-microbubbles, respectively, to further confirm specific targeting of scFvGPIIb/IIIa to activated platelets in the tumour stroma. Results Using scFvGPIIb/IIIa we successfully showed specific targeting of activated platelets within the microenvironment of human tumour xenografts models via three different molecular imaging modalities. The presence of platelets within the tumour microenvironment, and as such their relevance as a molecular target epitope in cancer was further confirmed via immunofluorescence of human tumour sections of various cancer types, thus validating the translational importance of our novel approach to human disease. Conclusion Our study provides proof of concept for imaging and localization of tumours by molecular targeting activated platelets. We illustrate the utility of a unique scFv as a versatile biotechnological tool which can be conjugated to various contrast agents for molecular imaging of cancer using three different imaging modalities. These findings warrant further development of this activated platelet specific scFvGPIIb/IIIa, potentially as a universal marker for cancer diagnosis and ultimately for drug delivery in an innovative theranostic approach.


Subject(s)
Blood Platelets/chemistry , Molecular Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/pathology , Optical Imaging/methods , Platelet Activation , Animals , Disease Models, Animal , Heterografts , Humans , Mice , Neoplasm Transplantation , Platelet Glycoprotein GPIIb-IIIa Complex/metabolism , Single-Chain Antibodies/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL