Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
J Dermatol Sci ; 109(3): 127-135, 2023 Mar.
Article in English | MEDLINE | ID: mdl-36966029

ABSTRACT

BACKGROUND: Antigen-stimulated naïve T cells differentiate into effector and memory T cells, of which resident memory T (TRM) cells reside permanently in organ tissues. Involvement of TRM cells has been indicated in pathological conditions of various skin diseases. CD122, which is the ß chain subunit of interleukin (IL)- 2 and IL-15 receptors, is expressed on immune cells including TRM cells. OBJECTIVE: To investigate whether CD122 signaling in skin CD8+ TRM cells mediates the development of mucocutaneous graft-versus-host disease (GVHD). METHODS: We used a genetically modified mouse expressing a membrane-bound form of chicken ovalbumin (OVA) under the control of the keratin 14 promoter, which develops GVHD-like erosive mucocutaneous disease resulting in sclerodermatous disease after transfer of OVA-specific T cell-receptor-transgenic CD8+ OT-I cells. Mice with mucocutaneous GVHD were treated with an anti-CD122 blocking antibody. RESULTS: Administration of an anti-CD122 blocking antibody suppresses the development of acute/chronic GVHD-like mucocutaneous disease in our murine model via the reduction of CD122-expressing memory CD8+ T cells, including skin, memory autoaggressive CD8+ T cells. Moreover, blockade of CD122, even after the establishment of acute GVHD, inhibited the development of chronic GVHD-like sclerodermatous disease via the reduction of epidermal and dermal TRM autoaggressive CD8+ T cells. CONCLUSION: Skin memory CD8+ T cells in particular mediate the development of mucocutaneous GVHD, and blockade of CD122 may be an effective treatment strategy, especially for sclerodermatous GVHD.


Subject(s)
Graft vs Host Disease , Scleroderma, Localized , Animals , Mice , CD8-Positive T-Lymphocytes , Graft vs Host Disease/prevention & control , Graft vs Host Disease/pathology , Memory T Cells , Mice, Inbred C57BL , Skin/pathology
2.
Sci Transl Med ; 10(450)2018 07 18.
Article in English | MEDLINE | ID: mdl-30021889

ABSTRACT

Vitiligo is an autoimmune disease of the skin mediated by CD8+ T cells that kill melanocytes and create white spots. Skin lesions in vitiligo frequently return after discontinuing conventional treatments, supporting the hypothesis that autoimmune memory is formed at these locations. We found that lesional T cells in mice and humans with vitiligo display a resident memory (TRM) phenotype, similar to those that provide rapid, localized protection against reinfection from skin and mucosal-tropic viruses. Interleukin-15 (IL-15)-deficient mice reportedly have impaired TRM formation, and IL-15 promotes TRM function ex vivo. We found that both human and mouse TRM express the CD122 subunit of the IL-15 receptor and that keratinocytes up-regulate CD215, the subunit required to display the cytokine on their surface to promote activation of T cells. Targeting IL-15 signaling with an anti-CD122 antibody reverses disease in mice with established vitiligo. Short-term treatment with anti-CD122 inhibits TRM production of interferon-γ (IFNγ), and long-term treatment depletes TRM from skin lesions. Short-term treatment with anti-CD122 can provide durable repigmentation when administered either systemically or locally in the skin. On the basis of these data, we propose that targeting CD122 may be a highly effective and even durable treatment strategy for vitiligo and other tissue-specific autoimmune diseases involving TRM.


Subject(s)
Antibodies, Blocking/therapeutic use , Interleukin-15/metabolism , Signal Transduction , Vitiligo/drug therapy , Vitiligo/immunology , Animals , Antibodies, Blocking/administration & dosage , Antibodies, Blocking/pharmacology , Antigens, CD/metabolism , CD8-Positive T-Lymphocytes/immunology , Disease Models, Animal , Epidermis/immunology , Humans , Immunologic Memory , Interferon-gamma/metabolism , Melanocytes/metabolism , Mice, Inbred C57BL , Phenotype , Receptors, Interleukin-15/metabolism , Vitiligo/pathology
3.
JCI Insight ; 3(2)2018 01 25.
Article in English | MEDLINE | ID: mdl-29367461

ABSTRACT

Signaling through IL-2/IL-15Rß (CD122) is essential for the differentiation and function of T cells and NK cells. A mAb against CD122 has been implicated to suppress autoimmune type 1 diabetes (T1D) development in animal models. However, the mechanisms remain poorly understood. We find that in vivo administration of an anti-CD122 mAb (CD122 blockade) restores immune tolerance in nonobese diabetic (NOD) mice via multiple mechanisms. First, CD122 blockade selectively ablates pathogenic NK cells and memory phenotype CD8+ T cells from pancreatic islets. In contrast, islet CD4+Foxp3+ Tregs are only mildly affected. Second, CD122 blockade suppresses IFN-γ production in islet immune cells. Third, CD122 blockade inhibits the conversion of islet Th17 cells into diabetogenic Th1 cells. Furthermore, a combination of anti-CD122 mAb and Treg-trophic cytokines (IL-2 or IL-33) enhances the abundance and function of islet Tregs. In summary, these data provide crucial mechanistic insights into CD122 blockade-mediated immunoregulation and support therapeutic benefits of this combinational treatment in T1D.


Subject(s)
Antibodies, Monoclonal/administration & dosage , Diabetes Mellitus, Type 1/drug therapy , Immune Tolerance/drug effects , Immunologic Factors/administration & dosage , Interleukin-2 Receptor beta Subunit/antagonists & inhibitors , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Disease Models, Animal , Drug Therapy, Combination/methods , Female , Hybridomas , Interleukin-2/administration & dosage , Interleukin-2 Receptor beta Subunit/immunology , Interleukin-33/administration & dosage , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Mice , Mice, Inbred NOD , Rats , Rats, Sprague-Dawley , Recombinant Proteins/administration & dosage , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Th1 Cells/drug effects , Th1 Cells/immunology , Th17 Cells/drug effects , Th17 Cells/immunology , Treatment Outcome
4.
Clin Cancer Res ; 14(9): 2775-84, 2008 May 01.
Article in English | MEDLINE | ID: mdl-18451245

ABSTRACT

PURPOSE: We generated a humanized antibody, HuLuc63, which specifically targets CS1 (CCND3 subset 1, CRACC, and SLAMF7), a cell surface glycoprotein not previously associated with multiple myeloma. To explore the therapeutic potential of HuLuc63 in multiple myeloma, we examined in detail the expression profile of CS1, the binding properties of HuLuc63 to normal and malignant cells, and the antimyeloma activity of HuLuc63 in preclinical models. EXPERIMENTAL DESIGN: CS1 was analyzed by gene expression profiling and immunohistochemistry of multiple myeloma samples and numerous normal tissues. HuLuc63-mediated antimyeloma activity was tested in vitro in antibody-dependent cellular cytotoxicity (ADCC) assays and in vivo using the human OPM2 xenograft model in mice. RESULTS: CS1 mRNA was expressed in >90% of 532 multiple myeloma cases, regardless of cytogenetic abnormalities. Anti-CS1 antibody staining of tissues showed strong staining of myeloma cells in all plasmacytomas and bone marrow biopsies. Flow cytometric analysis of patient samples using HuLuc63 showed specific staining of CD138+ myeloma cells, natural killer (NK), NK-like T cells, and CD8+ T cells, with no binding detected on hematopoietic CD34+ stem cells. HuLuc63 exhibited significant in vitro ADCC using primary myeloma cells as targets and both allogeneic and autologous NK cells as effectors. HuLuc63 exerted significant in vivo antitumor activity, which depended on efficient Fc-CD16 interaction as well as the presence of NK cells in the mice. CONCLUSIONS: These results suggest that HuLuc63 eliminates myeloma cells, at least in part, via NK-mediated ADCC and shows the therapeutic potential of targeting CS1 with HuLuc63 for the treatment of multiple myeloma.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Lymphocyte Subsets/metabolism , Multiple Myeloma/drug therapy , Plasma Cells/metabolism , Receptors, Immunologic/immunology , Receptors, Immunologic/metabolism , Animals , Antibodies, Monoclonal/immunology , Antibody-Dependent Cell Cytotoxicity , Cell Line, Tumor , Female , Gene Expression Profiling , Humans , Killer Cells, Natural/immunology , Lymphocyte Subsets/cytology , Mice , Mice, SCID , Multiple Myeloma/immunology , Plasma Cells/cytology , Receptors, Immunologic/genetics , Signaling Lymphocytic Activation Molecule Family , Xenograft Model Antitumor Assays
5.
J Immunol Methods ; 327(1-2): 40-52, 2007 Oct 31.
Article in English | MEDLINE | ID: mdl-17719061

ABSTRACT

We have developed a mammalian cell surface display vector, suitable for directly isolating IgG molecules based on their antigen-binding affinity and biological activity. Using an Epstein-Barr virus-derived episomal vector, antibody libraries are displayed as whole IgG molecules on the cell surface and screened for specific antigen binding by a combination of magnetic beads and fluorescence-activated cell sorting. Plasmids encoding antibodies with desired binding characteristics are recovered from sorted cells and are converted to the form for production of soluble IgG. Transiently expressed soluble IgG antibodies are individually tested for binding to target antigens, as well as for biological activities, such as neutralization. This vector system was used to generate antibody display libraries derived from spleen cDNA of chickens immunized with human and mouse IL-12. Chicken-human chimeric IgG1 antibodies that neutralize human and mouse IL-12 were successfully isolated from the library. The mammalian surface display vector developed in this work facilitates the isolation of monoclonal antibodies from essentially any species.


Subject(s)
Antibodies, Monoclonal/immunology , Immunoglobulin G/immunology , Interleukin-12/immunology , Peptide Library , Animals , Antibodies, Monoclonal/genetics , Antibody Affinity , Antibody Specificity , Biomedical Engineering , Cell Separation , Chickens , Flow Cytometry , Genetic Vectors , Humans , Immunoglobulin G/genetics , Mice
6.
Biotechnol Bioeng ; 96(2): 294-306, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-16897745

ABSTRACT

Presented is an antibody production platform based on the fed-batch culture of recombinant NS0-derived cell lines. NS0 host cells, obtained from the European Collection of Cell Cultures (ECACC, Salisbury, UK, Part No. 85110503), were first adapted to grow in a protein-free, cholesterol-free medium. The resulting host cell line was designated NS0-PFCF (protein-free, cholesterol-free). The five production cell lines presented here were generated using a common protocol consisting of transfection by electroporation and subcloning. The NS0-PFCF host cell line was transfected using a single expression vector containing the Escherichia coli xanthine-guanine phosphoribosyl transferase gene (gpt), and the antibody heavy and light chain genes driven by the CMV promoter. The five cell lines were chosen after one to three rounds of iterative subcloning, which resulted in a 19-64% increase in antibody productivity when four mother-daughter cell pairs were cultured in a fed-batch bioreactor process. The production cell lines were genetically characterized to determine antibody gene integrity, nucleotide sequences, copy number, and the number of insertion sites in the NS0 cell genome. Genetic characterization data indicate that each of the five production cell lines has a single stably integrated copy of the antibody expression vector, and that the antibody genes are correctly expressed. Stability of antibody production was evaluated for three of the five cell lines by comparing the early stage seed bank with the Working Cell Bank (WCB). Antibody productivity was shown to be stable in two of three cell lines evaluated, while one of the cell lines exhibited a 20% drop in productivity after passaging for approximately 4 weeks. These five NS0-derived production cell lines were successfully cultured to produce antibodies with acceptable product quality attributes in a standardized fed-batch bioreactor process, consistently achieving an average specific productivity of 20-60 pg/cell-day, and a volumetric productivity exceeding 120 mg/L-day (Burky et al., 2006). In contrast to the commonly available NS0 host cell line, which requires serum and cholesterol for growth, and the commonly used expression vector system, which uses a proprietary glutamine synthetase selection marker (GS-NS0), these NS0 cells are cholesterol-independent, grow well in a protein-free medium, use a non-proprietary selection marker, and do not require gene amplification for productivity improvement. These characteristics are advantageous for use of this NS0 cell line platform for manufacturing therapeutic antibodies.


Subject(s)
Antibodies, Monoclonal/biosynthesis , Cell Culture Techniques/methods , Animals , Antibodies, Monoclonal/immunology , Bioreactors , Cell Line , Cholesterol/pharmacology , Humans , Mice , Recombinant Proteins/biosynthesis , Recombinant Proteins/immunology
7.
J Immunol ; 176(1): 346-56, 2006 Jan 01.
Article in English | MEDLINE | ID: mdl-16365427

ABSTRACT

The serum half-life of IgG Abs is regulated by the neonatal Fc receptor (FcRn). By binding to FcRn in endosomes, IgG Abs are salvaged from lysosomal degradation and recycled to the circulation. Several studies have demonstrated a correlation between the binding affinity of IgG Abs to FcRn and their serum half-lives in mice, including engineered Ab fragments with longer serum half-lives. Our recent study extended this correlation to human IgG2 Ab variants in primates. In the current study, several human IgG1 mutants with increased binding affinity to human FcRn at pH 6.0 were generated that retained pH-dependent release. A pharmacokinetics study in rhesus monkeys of one of the IgG1 variants indicated that its serum half-life was approximately 2.5-fold longer than the wild-type Ab. Ag binding was unaffected by the Fc mutations, while several effector functions appeared to be minimally altered. These properties suggest that engineered Abs with longer serum half-lives may prove to be effective therapeutics in humans.


Subject(s)
Immunoglobulin G/blood , Models, Molecular , Protein Engineering , Animals , Antibody Affinity , Binding Sites, Antibody , Cytotoxicity, Immunologic , Half-Life , Histocompatibility Antigens Class I/immunology , Humans , Immunoglobulin G/chemistry , Macaca mulatta , Receptors, Fc/immunology
8.
Methods ; 36(1): 69-83, 2005 May.
Article in English | MEDLINE | ID: mdl-15848076

ABSTRACT

Since the introduction of hybridoma technology, monoclonal antibodies have become one of the most important tools in the biosciences, finding diverse applications including their use in the therapy of human disease. Initial attempts to use monoclonal antibodies as therapeutics were hampered, however, by the potent immunogenicity of mouse (and other rodent) antibodies in humans. Humanization technology has made it possible to remove the immunogenicity associated with the use of rodent antibodies, or at least to reduce it to an acceptable level for clinical use in humans, thus facilitating the application of monoclonal antibodies to the treatment of human disease. To date, nine humanized monoclonal antibodies have been approved for use as human therapeutics in the United States. In this paper, we describe procedures for antibody humanization with an emphasis on strategies for designing humanized antibodies with the aid of computer-guided modeling of antibody variable domains, using as an example the humanized anti-CD25 monoclonal antibody, Zenapax.


Subject(s)
Antibodies, Monoclonal/chemistry , Immunoglobulin G/chemistry , Protein Engineering/methods , Amino Acid Sequence , Animals , Antibodies, Monoclonal/biosynthesis , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Humanized , Base Sequence , Daclizumab , Humans , Immunoglobulin G/biosynthesis , Immunoglobulin G/genetics , Immunoglobulin Variable Region/genetics , Mice , Molecular Sequence Data , Protein Structure, Tertiary , Sequence Alignment
9.
Am J Pathol ; 166(4): 1009-16, 2005 Apr.
Article in English | MEDLINE | ID: mdl-15793282

ABSTRACT

Overexpression of amphiregulin has been shown to induce psoriasiform changes in the skin of transgenic mice shortly after birth. Therefore, amphiregulin has been suggested as a target for anti-psoriatic therapy. To test this theory, a humanized monoclonal antibody capable of neutralizing human amphiregulin was examined for anti-proliferative effects in the human skin-severe combined immunodeficient (SCID) mouse transplant model. The anti-amphiregulin antibody reduced epidermal thickness of transplanted psoriatic skin and also inhibited the hyperplastic response that developed in nonpsoriatic skin after transplantation. The same antibody also suppressed keratinocyte proliferation in monolayer culture in a dose-dependent manner. Under the same conditions in which keratinocyte proliferation was inhibited, the antibody had little effect on proliferation of human dermal fibroblasts and no effect on type I procollagen production by these cells. Taken together, these data indicate an important role for amphiregulin in psoriatic hyperplasia and suggest that inhibition of amphiregulin activity could be an efficacious therapeutic strategy for psoriasis. These data also suggest that the hyperplastic response occurring in nonpsoriatic human skin on transplantation to the SCID mouse is mediated, in large part, by amphiregulin.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Epidermis/drug effects , Glycoproteins/immunology , Hyperplasia/drug therapy , Intercellular Signaling Peptides and Proteins/immunology , Psoriasis/drug therapy , Skin Transplantation , Amphiregulin , Animals , Cells, Cultured , Dose-Response Relationship, Drug , EGF Family of Proteins , Epidermis/metabolism , Epidermis/pathology , Fibroblasts/drug effects , Fibroblasts/metabolism , Glycoproteins/metabolism , Glycoproteins/pharmacology , Humans , Hyperplasia/metabolism , Hyperplasia/pathology , Intercellular Signaling Peptides and Proteins/metabolism , Intercellular Signaling Peptides and Proteins/pharmacology , Keratinocytes/drug effects , Keratinocytes/metabolism , Mice , Mice, SCID , Psoriasis/genetics
10.
Appl Environ Microbiol ; 70(5): 2567-76, 2004 May.
Article in English | MEDLINE | ID: mdl-15128505

ABSTRACT

Two different humanized immunoglobulin G1(kappa) antibodies and an Fab' fragment were produced by Aspergillus niger. The antibodies were secreted into the culture supernatant. Both light and heavy chains were initially synthesized as fusion proteins with native glucoamylase. After antibody assembly, cleavage by A. niger KexB protease allowed the release of free antibody. Purification by hydrophobic charge induction chromatography proved effective at removing any antibody to which glucoamylase remained attached. Glycosylation at N297 in the Fc region of the heavy chain was observed, but this site was unoccupied on approximately 50% of the heavy chains. The glycan was of the high-mannose type, with some galactose present, and the size ranged from Hex(6)GlcNAc(2) to Hex(15)GlcNAc(2). An aglycosyl mutant form of antibody was also produced. No significant difference between the glycosylated antibody produced by Aspergillus and that produced by mammalian cell cultures was observed in tests for affinity, avidity, pharmacokinetics, or antibody-dependent cellular cytotoxicity function.


Subject(s)
Antibodies/metabolism , Aspergillus niger/genetics , Aspergillus niger/metabolism , Immunoglobulin Fab Fragments/metabolism , Immunoglobulin G/metabolism , Amino Acid Sequence , Animals , Cell Line , Glucan 1,4-alpha-Glucosidase/genetics , Glucan 1,4-alpha-Glucosidase/metabolism , Glycosylation , Humans , Molecular Sequence Data , Peptide Mapping , Rats , Recombinant Fusion Proteins/metabolism
11.
J Biol Chem ; 279(8): 6213-6, 2004 Feb 20.
Article in English | MEDLINE | ID: mdl-14699147

ABSTRACT

The neonatal Fc receptor (FcRn) plays an important role in regulating the serum half-lives of IgG antibodies. A correlation has been established between the pH-dependent binding affinity of IgG antibodies to FcRn and their serum half-lives in mice. In this study, molecular modeling was used to identify Fc positions near the FcRn binding site in a human IgG antibody that, when mutated, might alter the binding affinity of IgG to FcRn. Following mutagenesis, several IgG2 mutants with increased binding affinity to human FcRn at pH 6.0 were identified at Fc positions 250 and 428. These mutants do not bind to human FcRn at pH 7.5. A pharmacokinetics study of two mutant IgG2 antibodies with increased FcRn binding affinity indicated that they had serum half-lives in rhesus monkeys approximately 2-fold longer than the wild-type antibody.


Subject(s)
Immunoglobulin G/blood , Immunoglobulin G/chemistry , Animals , Antibodies/chemistry , Binding Sites , Binding Sites, Antibody , Binding, Competitive , Cell Line , Cloning, Molecular , DNA, Complementary/metabolism , Dose-Response Relationship, Drug , Half-Life , Histocompatibility Antigens Class I , Humans , Hydrogen-Ion Concentration , Immunoglobulin G/genetics , Immunoglobulin G/immunology , Inhibitory Concentration 50 , Kidney/cytology , Macaca mulatta , Models, Molecular , Mutagenesis , Mutation , Protein Binding , Receptors, Fc/chemistry , Time Factors
12.
J Immunol Methods ; 295(1-2): 9-19, 2004 Dec.
Article in English | MEDLINE | ID: mdl-15627607

ABSTRACT

Chicken anti-IL-12 monoclonal antibodies were isolated by phage display using spleen cells from a chicken immunized with human and mouse IL-12 as a source for library construction. One of the chicken monoclonal antibodies, DD2, exhibited binding to both human and mouse IL-12 in the single-chain Fv form and also after conversion to chicken-human chimeric IgG1/lambda antibody. The chicken DD2 variable regions were humanized by transferring their CDRs and several framework amino acids onto human acceptor variable regions. In the Vlambda, six chicken framework amino acids were identified to be important for the conformation of the CDR structure by computer modeling and therefore were retained in the humanized form; likewise, five chicken amino acids in the VH framework regions were retained in the humanized VH. The affinities of humanized DD2 IgG1/lambda to human and mouse IL-12 measured by competitive binding were nearly identical to those of chicken-human chimeric DD2 IgG1/lambda. This work demonstrates that humanization of chicken monoclonal antibodies assisted by computer modeling is possible, leading to a new way to generate therapeutic humanized antibodies against antigens to which the rodent immune system may fail to efficiently raise high affinity antibodies.


Subject(s)
Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/isolation & purification , Antibody Specificity/immunology , Chickens/immunology , Interleukin-12/immunology , Amino Acid Sequence , Animals , Antibodies, Monoclonal/chemistry , Antibody Affinity/immunology , Enzyme-Linked Immunosorbent Assay , Female , Humans , Immunoglobulin Variable Region/chemistry , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Models, Molecular , Molecular Sequence Data , Peptide Library
SELECTION OF CITATIONS
SEARCH DETAIL
...