Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Mol Cancer Ther ; 21(4): 607-615, 2022 04 01.
Article in English | MEDLINE | ID: mdl-35086952

ABSTRACT

Antibody-mediated tumor delivery of cytokines can overcome limitations of systemic administration (toxicity, short half-lives). Previous work showed improved antitumor potency of anti-CD20-IFNα fusion proteins in preclinical mouse models of B-cell lymphoma. Although tumor targeting is mediated by the antibody part of the fusion protein, the cytokine component might strongly influence biodistribution and pharmacokinetics, as a result of its affinity, size, valency, and receptor distribution. Here, we used immunoPET to study the in vivo biodistribution and tumor targeting of the anti-CD20 rituximab-murine IFNα1 fusion protein (Rit-mIFNα) and compared it with the parental mAb (rituximab, Rit). Rit-mIFNα and Rit were radiolabeled with zirconium-89 (89Zr, t1/2 78.4 hours) and injected into C3H mice bearing syngeneic B-cell lymphomas (38C13-hCD20). Dynamic [(2 hours post injection (p.i.)] and static (4, 24, and 72 hours) PET scans were acquired. Ex vivo biodistribution was performed after the final scan. Both 89Zr-Rit-mIFNα and 89Zr-Rit specifically target hCD20-expressing B-cell lymphoma in vivo. 89Zr-Rit-mIFNα showed specific uptake in tumors (7.6 ± 1.0 %ID/g at 75 hours p.i.), which was significantly lower than 89Zr-Rit (38.4 ± 9.9 %ID/g, P < 0.0001). ImmunoPET studies also revealed differences in the biodistribution, 89Zr-Rit-mIFNα showed rapid blood clearance and high accumulation in the liver compared with 89Zr-Rit. Importantly, immunoPET clearly revealed a therapeutic effect of the single 89Zr-Rit-mIFNα dose, resulting in smaller tumors and fewer lymph node metastases compared with mice receiving 89Zr-Rit. Mice receiving 89Zr-Rit-mIFNα had enlarged spleens, suggesting that systemic immune activation contributes to therapeutic efficacy in addition to the direct antitumoral activity of IFNα. In conclusion, immunoPET allows the noninvasive tracking and quantification of the antibody-cytokine fusion protein and helps understand the in vivo behavior and therapeutic efficacy.


Subject(s)
Lymphoma, B-Cell , Radioisotopes , Animals , Cell Line, Tumor , Humans , Lymphoma, B-Cell/drug therapy , Mice , Mice, Inbred C3H , Positron-Emission Tomography/methods , Radioisotopes/therapeutic use , Tissue Distribution , Zirconium/therapeutic use
2.
Immunity ; 51(2): 351-366.e6, 2019 08 20.
Article in English | MEDLINE | ID: mdl-31303400

ABSTRACT

Aging results in increased myelopoiesis, which is linked to the increased incidence of myeloid leukemias and production of myeloid-derived suppressor cells. Here, we examined the contribution of plasma cells (PCs) to age-related increases in myelopoiesis, as PCs exhibit immune regulatory function and sequester in bone marrow (BM). PC number was increased in old BM, and they exhibited high expression of genes encoding inflammatory cytokines and pathogen sensors. Antibody-mediated depletion of PCs from old mice reduced the number of myeloid-biased hematopoietic stem cells and mature myeloid cells to levels in young animals, but lymphopoiesis was not rejuvenated, indicating that redundant mechanisms inhibit that process. PCs also regulated the production of inflammatory factors from BM stromal cells, and disruption of the PC-stromal cell circuitry with inhibitors of the cytokines IL-1 and TNF-α attenuated myelopoiesis in old mice. Thus, the age-related increase in myelopoiesis is driven by an inflammatory network orchestrated by PCs.


Subject(s)
Aging/physiology , Bone Marrow/physiology , Hematopoietic Stem Cells/pathology , Inflammation/metabolism , Myelopoiesis/physiology , Plasma Cells/physiology , Animals , Cells, Cultured , Humans , Interleukin-1/metabolism , Lymphocyte Depletion , Mice , Mice, Inbred C57BL , Mice, Transgenic , Tumor Necrosis Factor-alpha/metabolism
3.
Cancer Res ; 79(10): 2748-2760, 2019 05 15.
Article in English | MEDLINE | ID: mdl-30885978

ABSTRACT

Although the majority of adult tissues express only hexokinase 1 (HK1) for glycolysis, most cancers express hexokinase 2 (HK2) and many coexpress HK1 and HK2. In contrast to HK1+HK2+ cancers, HK1-HK2+ cancer subsets are sensitive to cytostasis induced by HK2shRNA knockdown and are also sensitive to synthetic lethality in response to the combination of HK2shRNA knockdown, an oxidative phosphorylation (OXPHOS) inhibitor diphenyleneiodonium (DPI), and a fatty acid oxidation (FAO) inhibitor perhexiline (PER). The majority of human multiple myeloma cell lines are HK1-HK2+. Here we describe an antisense oligonucleotide (ASO) directed against human HK2 (HK2-ASO1), which suppressed HK2 expression in human multiple myeloma cell cultures and human multiple myeloma mouse xenograft models. The HK2-ASO1/DPI/PER triple-combination achieved synthetic lethality in multiple myeloma cells in culture and prevented HK1-HK2+ multiple myeloma tumor xenograft progression. DPI was replaceable by the FDA-approved OXPHOS inhibitor metformin (MET), both for synthetic lethality in culture and for inhibition of tumor xenograft progression. In addition, we used an ASO targeting murine HK2 (mHK2-ASO1) to validate the safety of mHK2-ASO1/MET/PER combination therapy in mice bearing murine multiple myeloma tumors. HK2-ASO1 is the first agent that shows selective HK2 inhibition and therapeutic efficacy in cell culture and in animal models, supporting clinical development of this synthetically lethal combination as a therapy for HK1-HK2+ multiple myeloma. SIGNIFICANCE: A first-in-class HK2 antisense oligonucleotide suppresses HK2 expression in cell culture and in in vivo, presenting an effective, tolerated combination therapy for preventing progression of HK1-HK2+ multiple myeloma tumors. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/79/10/2748/F1.large.jpg.


Subject(s)
Hexokinase/genetics , Multiple Myeloma/pathology , Oligonucleotides, Antisense/pharmacology , Synthetic Lethal Mutations , Animals , Cell Line, Tumor , Cell Proliferation , Humans , Mice , Xenograft Model Antitumor Assays
4.
J Interferon Cytokine Res ; 38(6): 239-254, 2018 06.
Article in English | MEDLINE | ID: mdl-29920129

ABSTRACT

An important emerging form of immunotherapy targeting B cell malignancies is chimeric antigen receptor (CAR) T cell therapy. Despite encouraging response rates of anti-CD19 CAR T cell therapy in B cell lymphomas, limited durability of response necessitates further study to potentiate CAR T cell efficacy. Antibody-targeted interferon (IFN) therapy is a novel approach in immunotherapy. Given the ability of IFNs to promote T cell activation and survival, target cell recognition, and cytotoxicity, we asked whether antibody-targeted IFN could enhance the antitumor effects of anti-CD19 CAR T cells. We produced an anti-CD20-IFN fusion protein containing the potent type 1 IFN isoform alpha14 (α14), and demonstrated its ability to suppress proliferation and induce apoptosis of human B cell lymphomas. Indeed, with the combination of anti-CD20-hIFNα14 and CAR T cells, we found enhanced cell killing among B cell lymphoma lines. Importantly, for all cell lines pretreated with anti-CD20-hIFNα14, the subsequent cytokine production by CAR T cells was markedly increased regardless of the degree of cell killing. Thus, several activities of CD19 CAR T cells were enhanced in the presence of anti-CD20-hIFNα14. These data suggest that antibody-targeted IFN may be an important novel approach to improving the efficacy of CAR T cell therapy.


Subject(s)
Antigens, CD19/immunology , Immunotherapy , Interferon-alpha/immunology , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/therapy , Receptors, Chimeric Antigen/immunology , T-Lymphocytes/immunology , Antigen-Antibody Reactions , Cell Line, Tumor , Humans
5.
Nat Biomed Eng ; 2(2): 124-137, 2018 02.
Article in English | MEDLINE | ID: mdl-31015629

ABSTRACT

As cells with aberrant force-generating phenotypes can directly lead to disease, cellular force-generation mechanisms are high-value targets for new therapies. Here, we show that single-cell force sensors embedded in elastomers enable single-cell force measurements with ~100-fold improvement in throughput than was previously possible. The microtechnology is scalable and seamlessly integrates with the multi-well plate format, enabling highly parallelized time-course studies. In this regard, we show that airway smooth muscle cells isolated from fatally asthmatic patients have innately greater and faster force-generation capacity in response to stimulation than healthy control cells. By simultaneously tracing agonist-induced calcium flux and contractility in the same cell, we show that the calcium level is ultimately a poor quantitative predictor of cellular force generation. Finally, by quantifying phagocytic forces in thousands of individual human macrophages, we show that force initiation is a digital response (rather than a proportional one) to the proper immunogen. By combining mechanobiology at the single-cell level with high-throughput capabilities, this microtechnology can support drug-discovery efforts for clinical conditions associated with aberrant cellular force generation.


Subject(s)
Elastomers/chemistry , Single-Cell Analysis/methods , Asthma/pathology , Cell Differentiation , Cells, Cultured , Fluorescent Dyes/chemistry , Formoterol Fumarate/pharmacology , Heterocyclic Compounds, 4 or More Rings/pharmacology , Humans , Macrophages/cytology , Macrophages/metabolism , Mesenchymal Stem Cells/cytology , Mesenchymal Stem Cells/metabolism , Microscopy, Fluorescence , Myocardial Contraction/drug effects , Myocytes, Smooth Muscle/cytology , Myocytes, Smooth Muscle/metabolism , Phagocytosis/drug effects
6.
Nat Biomed Eng ; 2(4): 265, 2018 Apr.
Article in English | MEDLINE | ID: mdl-31015734

ABSTRACT

In the version of this Article originally published, in Fig. 1a, all cells in the top schematic were missing, and in the bottom-left schematic showing multiple pattern shapes, two cells were missing in the bottom-right corner. This figure has now been updated in all versions of the Article.

7.
Oncoimmunology ; 6(3): e1290038, 2017.
Article in English | MEDLINE | ID: mdl-28405526

ABSTRACT

Interferon-α (IFNα) has multiple antitumor effects including direct antitumor toxicity and the ability to potently stimulate both innate and adaptive immunity. However, its clinical applications in the treatment of malignancies have been limited because of short half-life and serious adverse reactions when attempting to deliver therapeutically effective doses. To address these issues, we fused IFNα2a to the anti-vascular endothelial growth factor and receptor 2 (VEGFR2) antibody JZA00 with the goal of targeting it to the tumor microenvironment where it can stimulate the antitumor immune response. The fusion protein, JZA01, is effective against colorectal cancer by inhibiting angiogenesis, exhibiting direct cytotoxicity, and activating the antitumor immune response. Although JZA01 exhibited reduced IFNα2 activity in vitro compared with native IFNα2, VEGFR2 targeting permitted efficient antiproliferative, proapoptotic, antiangiogenesis, and immune-stimulating effects against the colorectal tumors HCT-116 and SW620. JZA01 showed in vivo efficacy in NOD-SCID mice-bearing established HCT-116 tumors. In conclusion, this study describes an antitumor immunotherapy that is highly promising for the treatment of colorectal cancer.

8.
MAbs ; 8(7): 1386-1397, 2016 10.
Article in English | MEDLINE | ID: mdl-27362935

ABSTRACT

Although recent advances have substantially improved the management of multiple myeloma, it remains an incurable malignancy. We now demonstrate that anti-CD138 molecules genetically fused to type I interferons (IFN) synergize with the approved therapeutic bortezomib in arresting the proliferation of human multiple myeloma cell lines both in vitro and in vivo. The anti-CD138-IFNα14 fusion protein was active in inducing increased expression of signal transducer and activator of transcription 1 (STAT1) and its phosphorylation while the cell death pathway induced by bortezomib included generation of reactive oxygen species. Interferon regulatory factor 4 (IRF4), an important survival factor for myeloma cells, was down regulated following combination treatment. Induction of cell death appeared to be caspase-independent because treatment with inhibitors of caspase activation did not decrease the level of cell death. The observed caspase-independent synergistic cell death involved mitochondrial membrane depolarization, and poly(ADP-ribose) polymerase-1 (PARP-1) cleavage, and resulted in enhanced induction of apoptosis. Importantly, using 2 different in vivo xenograft models, we found that combination therapy of anti-CD138-IFNα14 and bortezomib was able to cure animals with established tumors (7 of 8 using OCI-My5 or 8 of 8 using NCI-H929). Thus, the combination of anti-CD138-IFNα with bortezomib shows great promise as a novel therapeutic approach for the treatment of multiple myeloma, a malignancy for which there are currently no cures.


Subject(s)
Antineoplastic Agents/administration & dosage , Bortezomib/administration & dosage , Immunotherapy/methods , Interferon-alpha/administration & dosage , Multiple Myeloma , Syndecan-1/antagonists & inhibitors , Animals , Apoptosis/drug effects , Drug Synergism , Humans , Mice , Recombinant Fusion Proteins/administration & dosage , Signal Transduction/drug effects , Xenograft Model Antitumor Assays
9.
Int J Oncol ; 47(5): 1735-48, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26398317

ABSTRACT

Treatment of patients with B-NHL with rituximab and CHOP has resulted in significant clinical responses. However, a subset of patients develops resistance to further treatments. The mechanism of unresponsiveness in vivo is not known. We have reported the development of rituximab-resistant clones derived from B-NHL cell lines as models to investigate the mechanism of resistance. The resistant clones exhibit hyper-activated survival/anti-apoptotic pathways and no longer respond to a combination of rituximab and drugs. Recent studies reported the therapeutic efficacy in mice bearing B-cell lymphoma xenografts following treatment with the anti-CD20-hIFNα fusion protein. We hypothesized that the fusion protein may bypass rituximab resistance and inhibit survival signaling pathways. Treatment of the rituximab-resistant clones with anti-CD20-hIFNα, but not with rituximab, IFNα, or rituximab+IFNα resulted in significant inhibition of cell proliferation and induction of cell death. Treatment with anti-CD20-hIFNα sensitized the cells to apoptosis by CDDP, doxorubicin and Treanda. Treatment with anti-CD20-hIFNα inhibited the NF-κB and p38 MAPK activities and induced the activation of PKC-δ and Stat-1. These effects were corroborated by the use of the inhibitors SB203580 (p38 MAPK) and Rottlerin (PKC-δ). Treatment with SB203580 enhanced the sensitization of the resistant clone by anti-CD20-hIFNα to CDDP apoptosis. In contrast, treatment with Rotterin inhibited significantly the sensitization induced by anti-CD20-hIFNα. Overall, the findings demonstrate that treatment with anti-CD20-hIFNα reverses resistance of B-NHL. These findings suggest the potential application of anti-CD20-hIFNα in combination with drugs in patients unresponsive to rituximab-containing regimens.


Subject(s)
Antigens, CD20/genetics , Interferon-alpha/genetics , Lymphoma, B-Cell/drug therapy , Lymphoma, B-Cell/genetics , Oncogene Proteins, Fusion/genetics , Animals , Antigens, CD20/therapeutic use , Apoptosis , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Resistance, Neoplasm/genetics , Humans , Imidazoles/administration & dosage , Interferon-alpha/therapeutic use , Lymphoma, B-Cell/pathology , Mice , Pyridines/administration & dosage , Rituximab/administration & dosage , p38 Mitogen-Activated Protein Kinases
10.
Immunity ; 42(2): 379-390, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680277

ABSTRACT

Understanding how viruses subvert host immunity and persist is essential for developing strategies to eliminate infection. T cell exhaustion during chronic viral infection is well described, but effects on antibody-mediated effector activity are unclear. Herein, we show that increased amounts of immune complexes generated in mice persistently infected with lymphocytic choriomeningitis virus (LCMV) suppressed multiple Fcγ-receptor (FcγR) functions. The high amounts of immune complexes suppressed antibody-mediated cell depletion, therapeutic antibody-killing of LCMV infected cells and human CD20-expressing tumors, as well as reduced immune complex-mediated cross-presentation to T cells. Suppression of FcγR activity was not due to inhibitory FcγRs or high concentrations of free antibody, and proper FcγR functions were restored when persistently infected mice specifically lacked immune complexes. Thus, we identify a mechanism of immunosuppression during viral persistence with implications for understanding effective antibody activity aimed at pathogen control.


Subject(s)
Antibodies, Viral/immunology , Antigen-Antibody Complex/immunology , Immune Evasion/immunology , Lymphocytic Choriomeningitis/immunology , Receptors, IgG/antagonists & inhibitors , Animals , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antigens, CD20/biosynthesis , Antigens, CD20/immunology , B-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Immune Tolerance/immunology , Immunologic Factors/pharmacology , Lymphocyte Activation/immunology , Lymphocyte Depletion , Lymphocytic Choriomeningitis/virology , Lymphocytic choriomeningitis virus/immunology , Macrophages/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Phagocytosis/immunology , Receptors, IgG/immunology , Rituximab
11.
J Interferon Cytokine Res ; 35(4): 281-91, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25353626

ABSTRACT

Multiple myeloma (MM), a plasma cell malignancy, is the second most prevalent hematologic malignancy in the US. Although much effort has been made trying to understand the etiology and the complexities of this disease with the hope of developing effective therapies, MM remains incurable at this time. Because of their antiproliferative and proapoptotic activities, interferons (IFNs) have been used to treat various malignancies, including MM. Although some success has been observed, the inherent toxicities of IFNs limit their efficacy. To address this problem, we produced anti-CD138 antibody fusion proteins containing either IFNα2 or a mutant IFNα2 (IFNα2(YNS)) with the goal of targeting IFN to CD138-expressing cells, thereby achieving effective IFN concentrations at the site of the tumor in the absence of toxicity. The fusion proteins inhibited the proliferation and induced apoptosis of U266, ANBL-6, NCI-H929, and MM1-144 MM cell lines. The fusion proteins decreased the expression of IFN regulatory factor 4 (IRF4) in U266. In addition, the fusion proteins were effective against primary cells from MM patients, and treatment with fusion proteins prolonged survival in the U266 murine model of MM. These studies show that IFNα antibody fusion proteins can be effective novel therapeutics for the treatment of MM.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Interferons/therapeutic use , Multiple Myeloma/drug therapy , Syndecan-1/immunology , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Humans , Interferons/pharmacology , Mice , Signal Transduction
12.
Semin Oncol ; 41(5): 623-36, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25440607

ABSTRACT

The true potential of cytokine therapies in cancer treatment is limited by the inability to deliver optimal concentrations into tumor sites due to dose-limiting systemic toxicities. To maximize the efficacy of cytokine therapy, recombinant antibody-cytokine fusion proteins have been constructed by a number of groups to harness the tumor-targeting ability of monoclonal antibodies. The aim is to guide cytokines specifically to tumor sites where they might stimulate more optimal anti-tumor immune responses while avoiding the systemic toxicities of free cytokine therapy. Antibody-cytokine fusion proteins containing interleukin (IL)-2, IL-12, IL-21, tumor necrosis factor (TNF)α, and interferons (IFNs) α, ß, and γ have been constructed and have shown anti-tumor activity in preclinical and early-phase clinical studies. Future priorities for development of this technology include optimization of tumor targeting, bioactivity of the fused cytokine, and choice of appropriate agents for combination therapies. This review is intended to serve as a framework for engineering an ideal antibody-cytokine fusion protein, focusing on previously developed constructs and their clinical trial results.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Cytokines/therapeutic use , Immunotherapy/methods , Neoplasms/therapy , Recombinant Fusion Proteins/therapeutic use , Animals , Antibodies, Monoclonal/immunology , Cytokines/immunology , Humans , Neoplasms/immunology , Protein Engineering , Recombinant Fusion Proteins/immunology , Safety
13.
J Immunother ; 36(5): 305-18, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23719241

ABSTRACT

Type I interferons (IFNα/ß) are cytokines with a broad spectrum of antitumor activities including antiproliferative, proapoptotic, and immunostimulatory effects, and are potentially useful in the treatment of B-cell malignancies and other cancers. To improve antitumor potency and diminish the systemic side effects of IFN, we recently developed anti-CD20-IFNα fusion proteins with in vitro and in vivo efficacy against both mouse and human lymphomas expressing CD20. As IFNß binds more tightly to the IFNα/ß receptor (IFNAR) and has more potent antitumor activities, we have now constructed an anti-CD20 fusion protein with murine IFNß (mIFNß). Anti-CD20-mIFNß was more potent than recombinant mIFNß and anti-CD20-mIFNα in inhibiting the proliferation of a mouse B-cell lymphoma expressing human CD20 (38C13-huCD20). Growth inhibition was accompanied by caspase-independent apoptosis and DNA fragmentation. The efficacy of anti-CD20-mIFNß required the physical linkage of mIFNß to anti-CD20 antibody. Importantly, anti-CD20-mIFNß was active against tumor cells expressing low levels of IFNAR (38C13-huCD20 IFNAR). In vivo, established 38C13-huCD20 tumors were largely insensitive to rituximab or a nontargeted mIFNß fusion protein, yet treatment with anti-CD20-mIFNß eradicated 83% of tumors. Anti-CD20-mIFNß was also more potent in vivo against 38C13-huCD20 than anti-CD20-mIFNα, curing 75% versus 25% of tumors (P=0.001). Importantly, although anti-CD20-mIFNα could not eradicate 38C13-huCD20 IFNAR tumors, anti-CD20-mIFNß treatment prolonged survival (P=0.0003), and some animals remained tumor-free. Thus, antibody fusion proteins targeting mIFNß to tumors show promise as therapeutic agents, especially for use against tumors resistant to the effects of mIFNα.


Subject(s)
Antibodies, Monoclonal/pharmacology , Interferon-alpha/pharmacology , Interferon-beta/pharmacology , Lymphoma, B-Cell/therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Monoclonal, Murine-Derived/therapeutic use , Antibody Affinity/genetics , Antigens, CD20/immunology , Apoptosis/drug effects , Cell Growth Processes/drug effects , Cell Line, Tumor , Drug Resistance, Neoplasm , Interferon-alpha/therapeutic use , Interferon-beta/genetics , Interferon-beta/metabolism , Lymphoma, B-Cell/immunology , Mice , Receptor, Interferon alpha-beta/metabolism , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Rituximab
14.
J Biol Chem ; 288(15): 10395-405, 2013 Apr 12.
Article in English | MEDLINE | ID: mdl-23439650

ABSTRACT

A critical step in the induction of adaptive mucosal immunity is antigen transcytosis, in which luminal antigens are transported to organized lymphoid tissues across the follicle-associated epithelium (FAE) of Peyer's patches. However, virtually nothing is known about intracellular signaling proteins and transcription factors that regulate apical-to-basolateral transcytosis. The FAE can transcytose a variety of luminal contents, including inert particles, in the absence of specific opsonins. Furthermore, it expresses receptors for secretory immunoglobulin A (SIgA), the main antibody in mucosal secretions, and uses them to efficiently transcytose SIgA-opsonized particles present in the lumen. Using a human FAE model, we show that the tyrosine kinase HCK regulates apical-to-basolateral transcytosis of non-opsonized and SIgA-opsonized particles. We also show that, in cultured intestinal epithelial cells, ectopic expression of the transcription factor SPIB or EHF is sufficient to activate HCK-dependent apical-to-basolateral transcytosis of these particles. Our results provide the first molecular insights into the intracellular regulation of antigen sampling at mucosal surfaces.


Subject(s)
DNA-Binding Proteins/immunology , Intestinal Mucosa/immunology , Models, Immunological , Peyer's Patches/immunology , Proto-Oncogene Proteins c-hck/immunology , Transcription Factors/immunology , Transcytosis/immunology , Animals , Cell Line , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Humans , Immunoglobulin A/immunology , Immunoglobulin A/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Mice , Peyer's Patches/cytology , Peyer's Patches/metabolism , Proto-Oncogene Proteins c-hck/genetics , Proto-Oncogene Proteins c-hck/metabolism , Transcription Factors/genetics , Transcription Factors/metabolism
15.
J Transl Med ; 11: 23, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23360526

ABSTRACT

BACKGROUND: Chimeric Antigen Receptors (CARs) consist of the antigen-recognition portion of a monoclonal antibody fused to an intracellular signaling domain capable of activating T-cells. CARs displayed on the surface of transduced cells perform non-MHC-restricted antigen recognition and activating intracellular signaling pathways for induction of target cytolysis, cytokine secretion and proliferation. Clinical trials are in progress assessing the use of mature T-lymphocytes transduced with CARs targeting CD19 antigen to treat B-lineage malignancies. CD19 is an attractive target for immunotherapy because of its consistent and specific expression in most of the stages of maturation and malignancies of B-lymphocyte origin, but not on hematopoietic stem cells. Antibodies against the extracellular domain of the CAR molecule (anti-Fab, Fc or idiotype) have been used for detection of CAR expression in research and clinical samples by flow cytometry, but may need development for each construct and present significant background in samples from xenograft models. METHODS: A specific reagent for the detection of anti-CD19 CAR expression was developed, a fusion protein consisting of human CD19 extracellular domains and the Fc region of human IgG1 (CD19sIg). Genes encoding CD19sIg fusion proteins were constructed by fusing either exons 1 to 3 (CD19sIg1-3) or exons 1 to 4 (CD19sIg1-4) of the human CD19 cDNA to a human IgG1Fc fragment. These fusion proteins are intended to work in similar fashion as the MHC Tetramers used for identification of antigen-specific T-cells, and may also have other applications in studies of activation of anti-CD19 CAR bearing cells. The CD19sIg proteins were produced from 293 T cells by stable lentiviral vector transduction and purification from culture medium. RESULTS: ELISA assays using several different monoclonal antibodies to CD19 demonstrated dose-related specific binding by the fusion molecule CD19sIg1-4, but no binding by CD19sIg1-3. Conjugation of the CD19sIg1-4 fusion protein to Alexa Fluor 488 allowed specific and sensitive staining of anti-CD19 CAR-bearing cells for flow cytometry assays, detecting as low as 0.5% of CAR-modified primary cells with minimal background staining. CONCLUSIONS: This fusion molecule is a sensitive reagent for detection of anti-CD19 CAR derived from any monoclonal antibody present in CAR-modified T-cells.


Subject(s)
Antigens, CD19/chemistry , Immunoglobulin Fragments/chemistry , Receptors, Antigen/chemistry , Recombinant Fusion Proteins/metabolism , Animals , Antibodies, Monoclonal/chemistry , Cell Proliferation , Cell Transplantation , Dose-Response Relationship, Drug , Enzyme-Linked Immunosorbent Assay , Flow Cytometry , HEK293 Cells , Humans , Immunoglobulin G/chemistry , Leukocytes, Mononuclear/cytology , Lymphocyte Activation , Mice , Mice, Inbred NOD , Mice, SCID , Protein Structure, Tertiary , Recombinant Fusion Proteins/chemistry , T-Lymphocytes/cytology
16.
Antimicrob Agents Chemother ; 55(11): 5090-8, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21876045

ABSTRACT

Emergence of drug-resistant mutant viruses during the course of antiretroviral therapy is a major hurdle that limits the success of chemotherapeutic treatment to suppress human immunodeficiency virus type 1 (HIV-1) replication and AIDS progression. Development of new drugs and careful patient management based on resistance genotyping data are important for enhancing therapeutic efficacy. However, identifying changes leading to drug resistance can take years of clinical studies, and conventional in vitro assays are limited in generating reliable drug resistance data. Here we present an efficient in vitro screening assay for selecting drug-resistant variants from a library of randomly mutated HIV-1 strains generated by transposon-directed base-exchange mutagenesis. As a test of principle, we screened a library of mutant HIV-1 strains containing random mutations in the protease gene by using a reporter T-cell line in the presence of the protease inhibitor (PI) nelfinavir (NFV). Analysis of replicating viruses from a single round of infection identified 50 amino acid substitutions at 35 HIV-1 protease residue positions. The selected mutant viruses showed specific resistance to NFV and included most of the known NFV resistance mutations. Therefore, the new assay is efficient for identifying changes leading to drug resistance. The data also provide insights into the molecular mechanisms underlying the development of drug resistance.


Subject(s)
HIV Protease Inhibitors/pharmacology , HIV-1/drug effects , HIV-1/genetics , Cell Line , Drug Resistance, Viral/genetics , Humans , Mutation , Nelfinavir/pharmacology
17.
Mol Immunol ; 48(15-16): 2009-18, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21723612

ABSTRACT

Complement can be activated via three pathways: classical, alternative, and lectin. Cryptococcus gattii and Cryptococcus neoformans are closely related fungal pathogens possessing a polysaccharide capsule composed mainly of glucuronoxylomannan (GXM), which serves as a site for complement activation and deposition of complement components. We determined C3 deposition on Cryptococcus spp. by flow cytometry and confocal microscopy after incubation with serum from C57BL/6J mice as well as mice deficient in complement components C4, C3, factor B, and mannose binding lectin (MBL). C. gattii and C. neoformans activate complement in EGTA-treated serum indicating that they can activate the alternative pathway. However, complement activation was seen with factor B(-/-) serum suggesting activation could also take place in the absence of a functional alternative pathway. Furthermore, we uncovered a role for C4 in the alternative pathway activation by Cryptococcus spp. We also identified an unexpected and complex role for MBL in complement activation by Cryptococcus spp. No complement activation occurred in the absence of MBL-A and -C proteins although activation took place when the lectin binding activity of MBL was disrupted by calcium chelation. In addition, alternative pathway activation by C. neoformans required both MBL-A and -C, while either MBL-A or -C was sufficient for alternative pathway activation by C. gattii. Thus, complement activation by Cryptococcus spp. can take place through multiple pathways and complement activation via the alternative pathway requires the presence of C4 and MBL proteins.


Subject(s)
Antigens, Fungal/immunology , Complement C3/immunology , Complement Pathway, Alternative/immunology , Complement Pathway, Mannose-Binding Lectin/immunology , Cryptococcus/immunology , Animals , Cell Separation , Complement C4/immunology , Complement Pathway, Classical/immunology , Flow Cytometry , Mannose-Binding Lectin/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Microscopy, Confocal , Polysaccharides/immunology
18.
Mol Immunol ; 48(15-16): 1818-26, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21632111

ABSTRACT

The Fcα/µ receptor (Fcα/µR) is an unusual Fc receptor in that it binds to two different antibody isotypes, IgA and IgM. This receptor is of interest because it is thought to be involved in the capture of IgA- and IgM-immune complexes and antigen presentation. To further characterize this receptor, we were able to stably express human Fcα/µR on the surface of the 293T cell line. Using this system, we determined the affinity of the interactions of the receptor with IgA and IgM, which led to novel insights including the important finding that IgM polymers can bind to human Fcα/µR in the absence of J chain. This is in contrast to the polymeric immunoglobulin receptor (pIgR), which requires the presence of J chain to bind to polymeric IgA and IgM. The dissociation constants (K(d)) of all of the different human IgA isotypes and allotypes for human Fcα/µR were determined, and we show that the N-linked glycans on IgA1 are not required for binding to the receptor. In addition, we demonstrate that IgA can be rapidly internalized by human Fcα/µR in the presence of cross-linking antibody.


Subject(s)
Immunoglobulin A/metabolism , Immunoglobulin M/metabolism , Receptors, Fc/metabolism , Cell Line , Cell Separation , Flow Cytometry , Humans , Immunoglobulin A/chemistry , Immunoglobulin M/chemistry , Transfection
19.
MAbs ; 2(3): 320-34, 2010.
Article in English | MEDLINE | ID: mdl-20431350

ABSTRACT

The development and production of recombinant monoclonal antibodies is well established. Although most of these are IgGs, there is also great interest in producing recombinant IgAs since this isotype plays a critical role in providing immunologic protection at mucosal surfaces. The choice of expression system for production of recombinant antibodies is crucial because they are glycoproteins containing at least one N-linked carbohydrate. These glycans have been shown to contribute to the stability, pharmacokinetics and biologic function of antibodies. We have produced recombinant human IgA1 and all three allotypes of IgA2 in murine myeloma and CHO cell lines to systematically characterize and compare the N-linked glycans. Recombinant IgAs produced in murine myelomas differ significantly from IgA found in humans in that they contain the highly immunogenic Galalpha(1,3)Gal epitope and N-glycolylneuraminic acid residues, indicating that murine myeloma is not the optimal expression system for the production of human IgA. In contrast, IgAs produced in CHO cells contained glycans that were more similar to those found on human IgA. Expression of IgA1 and IgA2 in Lec2 and Lec8 cell lines that are defective in glycan processing resulted in a less complex pool of N-glycans. In addition, the level of sialylation of rIgAs produced in murine and CHO cells was significantly lower than that previously reported for serum IgA1. These data underscore the importance of choosing the appropriate cell line for the production of glycoproteins with therapeutic potential.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/chemistry , Immunoglobulin A/biosynthesis , Immunoglobulin A/chemistry , Polysaccharides/chemistry , Animals , Antibodies, Monoclonal/genetics , CHO Cells , Cell Line, Tumor , Cricetinae , Cricetulus , Immunoglobulin A/genetics , Mice , Protein Engineering , Recombinant Proteins/biosynthesis , Recombinant Proteins/chemistry , Recombinant Proteins/genetics
20.
Blood ; 115(14): 2864-71, 2010 Apr 08.
Article in English | MEDLINE | ID: mdl-20139095

ABSTRACT

The anti-CD20 antibody rituximab has substantially improved outcomes in patients with B-cell non-Hodgkin lymphomas. However, many patients are not cured by rituximab-based therapies, and overcoming de novo or acquired rituximab resistance remains an important challenge to successful treatment of B-cell malignancies. Interferon-alpha (IFNalpha) has potent immunostimulatory properties and antiproliferative effects against some B-cell cancers, but its clinical utility is limited by systemic toxicity. To improve the efficacy of CD20-targeted therapy, we constructed fusion proteins consisting of anti-CD20 and murine or human IFNalpha. Fusion proteins had reduced IFNalpha activity in vitro compared with native IFNalpha, but CD20 targeting permitted efficient antiproliferative and proapoptotic effects against an aggressive rituximab-insensitive human CD20(+) murine lymphoma (38C13-huCD20) and a human B-cell lymphoma (Daudi). In vivo efficacy was demonstrated against established 38C13-huCD20 grown in syngeneic immunocompetent mice and large, established Daudi xenografts grown in nude mice. Optimal tumor eradication required CD20 targeting, with 87% of mice cured of rituximab-insensitive tumors. Gene knockdown studies revealed that tumor eradication required expression of type I IFN receptors on the tumor cell surface. Targeting type I IFNs to sites of B-cell lymphoma by fusion to anti-CD20 antibodies represents a potentially useful strategy for treatment of B-cell malignancies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Interferon-alpha/pharmacology , Lymphoma, B-Cell/drug therapy , Recombinant Fusion Proteins/pharmacology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Murine-Derived , Antiviral Agents/pharmacology , Cell Line, Tumor , Drug Delivery Systems , Gene Knockdown Techniques , Humans , Interferon-alpha/genetics , Mice , Mice, Nude , Receptors, Interferon , Recombinant Fusion Proteins/genetics , Rituximab , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...