Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 38
Filter
Add more filters










Publication year range
1.
Oncoimmunology ; 11(1): 2113697, 2022.
Article in English | MEDLINE | ID: mdl-36016696

ABSTRACT

The use of T-cell engagers (TCEs) to treat solid tumors is challenging, and several have been limited by narrow therapeutic windows due to substantial on-target, off-tumor toxicities due to the expression of low levels of target antigens on healthy tissues. Here, we describe TNB-928B, a fully human TCE that has a bivalent binding arm for folate receptor alpha (FRα) to selectively target FRα overexpressing tumor cells while avoiding the lysis of cells with low levels of FRα expression. The bivalent design of the FRα binding arm confers tumor selectivity due to low-affinity but high-avidity binding to high FRα antigen density cells. TNB-928B induces preferential effector T-cell activation, proliferation, and selective cytotoxic activity on high FRα expressing cells while sparing low FRα expressing cells. In addition, TNB-928B induces minimal cytokine release compared to a positive control TCE containing OKT3. Moreover, TNB-928B exhibits substantial ex vivo tumor cell lysis using endogenous T-cells and robust tumor clearance in vivo, promoting T-cell infiltration and antitumor activity in mouse models of ovarian cancer. TNB-928B exhibits pharmacokinetics similar to conventional antibodies, which are projected to enable favorable administration in humans. TNB-928B is a novel TCE with enhanced safety and specificity for the treatment of ovarian cancer.


Subject(s)
Antibodies, Bispecific , Ovarian Neoplasms , Animals , Antibodies, Bispecific/therapeutic use , Carcinoma, Ovarian Epithelial , Female , Folate Receptor 1/metabolism , Folate Receptor 1/therapeutic use , Humans , Mice , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , T-Lymphocytes
2.
MAbs ; 14(1): 2095949, 2022.
Article in English | MEDLINE | ID: mdl-35867844

ABSTRACT

Cluster of differentiation 38 (CD38) is an ecto-enzyme expressed primarily on immune cells that metabolize nicotinamide adenine dinucleotide (NAD+) to adenosine diphosphate ribose or cyclic ADP-ribose and nicotinamide. Other substrates of CD38 include nicotinamide adenine dinucleotide phosphate and nicotinamide mononucleotide, a critical NAD+ precursor in the salvage pathway. NAD+ is an important coenzyme involved in several metabolic pathways and is a required cofactor for the function of sirtuins (SIRTs) and poly (adenosine diphosphate-ribose) polymerases. Declines in NAD+ levels are associated with metabolic and inflammatory diseases, aging, and neurodegenerative disorders. To inhibit CD38 enzyme activity and boost NAD+ levels, we developed TNB-738, an anti-CD38 biparatopic antibody that pairs two non-competing heavy chain-only antibodies in a bispecific format. By simultaneously binding two distinct epitopes on CD38, TNB-738 potently inhibited its enzymatic activity, which in turn boosted intracellular NAD+ levels and SIRT activities. Due to its silenced IgG4 Fc, TNB-738 did not deplete CD38-expressing cells, in contrast to the clinically available anti-CD38 antibodies, daratumumab, and isatuximab. TNB-738 offers numerous advantages compared to other NAD-boosting therapeutics, including small molecules, and supplements, due to its long half-life, specificity, safety profile, and activity. Overall, TNB-738 represents a novel treatment with broad therapeutic potential for metabolic and inflammatory diseases associated with NAD+ deficiencies.Abbreviations: 7-AAD: 7-aminoactinomycin D; ADCC: antibody dependent cell-mediated cytotoxicity; ADCP: antibody dependent cell-mediated phagocytosis; ADPR: adenosine diphosphate ribose; APC: allophycocyanin; cADPR: cyclic ADP-ribose; cDNA: complementary DNA; BSA: bovine serum albumin; CD38: cluster of differentiation 38; CDC: complement dependent cytotoxicity; CFA: Freund's complete adjuvant; CHO: Chinese hamster ovary; CCP4: collaborative computational project, number 4; COOT: crystallographic object-oriented toolkit; DAPI: 4',6-diamidino-2-phenylindole; DNA: deoxyribonucleic acid; DSC: differential scanning calorimetry; 3D: three dimensional; εNAD+: nicotinamide 1,N6-ethenoadenine dinucleotide; ECD: extracellular domain; EGF: epidermal growth factor; FACS: fluorescence activated cell sorting; FcγR: Fc gamma receptors; FITC: fluorescein isothiocyanate; HEK: human embryonic kidney; HEPES: 4-(2-hydroxyethyl)-1-piperazineethanesulfonic acid; IgG: immunoglobulin; IFA: incomplete Freund's adjuvant; IFNγ: Interferon gamma; KB: kinetic buffer; kDa: kilodalton; KEGG: kyoto encyclopedia of genes and genomes; LDH: lactate dehydrogenase; M: molar; mM: millimolar; MFI: mean fluorescent intensity; NA: nicotinic acid; NAD: nicotinamide adenine dinucleotide; NADP: nicotinamide adenine dinucleotide phosphate; NAM: nicotinamide; NGS: next-generation sequencing; NHS/EDC: N-Hydroxysuccinimide/ ethyl (dimethylamino propyl) carbodiimide; Ni-NTA: nickel-nitrilotriacetic acid; nL: nanoliter; NK: natural killer; NMN: nicotinamide mononucleotide; OD: optical density; PARP: poly (adenosine diphosphate-ribose) polymerase; PBS: phosphate-buffered saline; PBMC: peripheral blood mononuclear cell; PDB: protein data bank; PE: phycoerythrin; PISA: protein interfaces, surfaces, and assemblies: PK: pharmacokinetics; mol: picomolar; RNA: ribonucleic acid; RLU: relative luminescence units; rpm: rotations per minute; RU: resonance unit; SEC: size exclusion chromatography; SEM: standard error of the mean; SIRT: sirtuins; SPR: surface plasmon resonance; µg: microgram; µM: micromolar; µL: microliter.


Subject(s)
NAD , Sirtuins , Adenosine Diphosphate Ribose/chemistry , Adenosine Diphosphate Ribose/metabolism , Animals , CHO Cells , Cricetinae , Cricetulus , Cyclic ADP-Ribose , Humans , Immunoglobulin G , Leukocytes, Mononuclear/metabolism , NAD/chemistry , NAD/metabolism , NADP , Niacinamide , Nicotinamide Mononucleotide , Ribose
3.
J Immunother Cancer ; 9(6)2021 06.
Article in English | MEDLINE | ID: mdl-34088740

ABSTRACT

BACKGROUND: Therapeutic options currently available for metastatic castration-resistant prostate cancer (mCRPC) do not extend median overall survival >6 months. Therefore, the development of novel and effective therapies for mCRPC represents an urgent medical need. T cell engagers (TCEs) have emerged as a promising approach for the treatment of mCRPC due to their targeted mechanism of action. However, challenges remain in the clinic due to the limited efficacy of TCEs observed thus far in solid tumors as well as the toxicities associated with cytokine release syndrome (CRS) due to the usage of high-affinity anti-CD3 moieties such as OKT3. METHODS: Using genetically engineered transgenic rats (UniRat and OmniFlic) that express fully human IgG antibodies together with an NGS-based antibody discovery pipeline, we developed TNB-585, an anti-CD3xPSMA TCE for the treatment of mCRPC. TNB-585 pairs a tumor-targeting anti-PSMA arm together with a unique, low-affinity anti-CD3 arm in bispecific format. We tested TNB-585 in T cell-redirected cytotoxicity assays against PSMA+ tumor cells in both two-dimensional (2D) cultures and three-dimensional (3D) spheroids as well as against patient-derived prostate tumor cells. Cytokines were measured in culture supernatants to assess the ability of TNB-585 to induce tumor killing with low cytokine release. TNB-585-mediated T cell activation, proliferation, and cytotoxic granule formation were measured to investigate the mechanism of action. Additionally, TNB-585 efficacy was evaluated in vivo against C4-2 tumor-bearing NCG mice. RESULTS: In vitro, TNB-585 induced activation and proliferation of human T cells resulting in the killing of PSMA+ prostate tumor cells in both 2D cultures and 3D spheroids with minimal cytokine release and reduced regulatory T cell activation compared with a positive control antibody that contains the same anti-PSMA arm but a higher affinity anti-CD3 arm (comparable with OKT3). In addition, TNB-585 demonstrated potent efficacy against patient-derived prostate tumors ex vivo and induced immune cell infiltration and dose-dependent tumor regression in vivo. CONCLUSIONS: Our data suggest that TNB-585, with its low-affinity anti-CD3, may be efficacious while inducing a lower incidence and severity of CRS in patients with prostate cancer compared with TCEs that incorporate high-affinity anti-CD3 domains.


Subject(s)
Antibodies, Bispecific/administration & dosage , Antigens, Surface/immunology , CD3 Complex/immunology , Glutamate Carboxypeptidase II/immunology , Immunoglobulin G/genetics , Prostatic Neoplasms, Castration-Resistant/drug therapy , Animals , Antibodies, Bispecific/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Dose-Response Relationship, Drug , Humans , Macaca fascicularis , Male , Mice , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/immunology , Rats , Rats, Transgenic , Xenograft Model Antitumor Assays
4.
Sci Rep ; 11(1): 10592, 2021 05 19.
Article in English | MEDLINE | ID: mdl-34011961

ABSTRACT

The use of recombinant interleukin-2 (IL-2) as a therapeutic protein has been limited by significant toxicities despite its demonstrated ability to induce durable tumor-regression in cancer patients. The adverse events and limited efficacy of IL-2 treatment are due to the preferential binding of IL-2 to cells that express the high-affinity, trimeric receptor, IL-2Rαßγ such as endothelial cells and T-regulatory cells, respectively. Here, we describe a novel bispecific heavy-chain only antibody which binds to and activates signaling through the heterodimeric IL-2Rßγ receptor complex that is expressed on resting T-cells and NK cells. By avoiding binding to IL-2Rα, this molecule circumvents the preferential T-reg activation of native IL-2, while maintaining the robust stimulatory effects on T-cells and NK-cells in vitro. In vivo studies in both mice and cynomolgus monkeys confirm the molecule's in vivo biological activity, extended pharmacodynamics due to the Fc portion of the molecule, and enhanced safety profile. Together, these results demonstrate that the bispecific antibody is a safe and effective IL-2R agonist that harnesses the benefits of the IL-2 signaling pathway as a potential anti-cancer therapy.


Subject(s)
Antibodies, Bispecific/pharmacology , Antineoplastic Agents, Immunological/pharmacology , Interleukin Receptor Common gamma Subunit/agonists , Interleukin-2 Receptor beta Subunit/agonists , Lymphocytes/drug effects , Animals , CHO Cells , Cricetulus , Drug Evaluation, Preclinical , HEK293 Cells , Humans , Interleukin Receptor Common gamma Subunit/immunology , Interleukin-2 Receptor beta Subunit/immunology , Macaca fascicularis , Male , Mice, Inbred BALB C
5.
MAbs ; 13(1): 1890411, 2021.
Article in English | MEDLINE | ID: mdl-33818299

ABSTRACT

The therapeutic potential of targeting CD19 in B cell malignancies has garnered attention in the past decade, resulting in the introduction of novel immunotherapy agents. Encouraging clinical data have been reported for T cell-based targeting agents, such as anti-CD19/CD3 bispecific T-cell engager blinatumomab and chimeric antigen receptor (CAR)-T therapies, for acute lymphoblastic leukemia and B cell non-Hodgkin lymphoma (B-NHL). However, clinical use of both blinatumomab and CAR-T therapies has been limited due to unfavorable pharmacokinetics (PK), significant toxicity associated with cytokine release syndrome and neurotoxicity, and manufacturing challenges. We present here a fully human CD19xCD3 bispecific antibody (TNB-486) for the treatment of B-NHL that could address the limitations of the current approved treatments. In the presence of CD19+ target cells and T cells, TNB-486 induces tumor cell lysis with minimal cytokine release, when compared to a positive control. In vivo, TNB-486 clears CD19+ tumor cells in immunocompromised mice in the presence of human peripheral blood mononuclear cells in multiple models. Additionally, the PK of TNB-486 in mice or cynomolgus monkeys is similar to conventional antibodies. This new T cell engaging bispecific antibody targeting CD19 represents a novel therapeutic that induces potent T cell-mediated tumor-cell cytotoxicity uncoupled from high levels of cytokine release, making it an attractive candidate for B-NHL therapy.


Subject(s)
Antibodies, Bispecific/pharmacology , Antibodies, Monoclonal, Humanized/pharmacology , Antineoplastic Agents, Immunological/pharmacology , CD4-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/drug effects , Cytokines/metabolism , Cytotoxicity, Immunologic/drug effects , Lymphocyte Activation/drug effects , Lymphoma, Non-Hodgkin/drug therapy , Animals , Antibodies, Bispecific/pharmacokinetics , Antibodies, Monoclonal, Humanized/pharmacokinetics , Antigens, CD19/immunology , Antineoplastic Agents, Immunological/pharmacokinetics , CD3 Complex/antagonists & inhibitors , CD3 Complex/immunology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Coculture Techniques , Humans , K562 Cells , Lymphoma, Non-Hodgkin/immunology , Lymphoma, Non-Hodgkin/metabolism , Macaca fascicularis , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Xenograft Model Antitumor Assays
6.
MAbs ; 12(1): 1846900, 2020.
Article in English | MEDLINE | ID: mdl-33228444

ABSTRACT

Transgenic animals incorporating human antibody genes are extremely attractive for drug development because they obviate subsequent antibody humanization procedures required for therapeutic translation. Transgenic platforms have previously been established using mice, but also more recently rats, chickens, and cows and are now in abundant use for drug development. However, rabbit-based antibody generation, with a strong track record for specificity and affinity, is able to include gene conversion mediated sequence diversification, thereby enhancing binder maturation and improving the variance/selection of output antibodies in a different way than in rodents. Since it additionally frequently permits good binder generation against antigens that are only weakly immunogenic in other organisms, it is a highly interesting species for therapeutic antibody generation. We report here on the generation, utilization, and analysis of the first transgenic rabbit strain for human antibody production. Through the knockout of endogenous IgM genes and the introduction of human immunoglobulin sequences, this rabbit strain has been engineered to generate a highly diverse human IgG antibody repertoire. We further incorporated human CD79a/b and Bcl2 (B-cell lymphoma 2) genes, which enhance B-cell receptor expression and B-cell survival. Following immunization against the angiogenic factor BMP9 (Bone Morphogenetic Proteins 9), we were able to isolate a set of exquisitely affine and specific neutralizing antibodies from these rabbits. Sequence analysis of these binders revealed that both somatic hypermutation and gene conversion are fully operational in this strain, without compromising the very high degree of humanness. This powerful new transgenic strategy will allow further expansion of the use of endogenous immune mechanisms in drug development.


Subject(s)
Animals, Genetically Modified , Antibody Affinity/immunology , Antibody Specificity/immunology , B-Lymphocytes/immunology , Immunoglobulin G/immunology , Animals , Humans , Immunoglobulin G/genetics , Rabbits
7.
MAbs ; 11(4): 639-652, 2019.
Article in English | MEDLINE | ID: mdl-30698484

ABSTRACT

T-cell-recruiting bispecific antibodies (T-BsAbs) have shown potent tumor killing activity in humans, but cytokine release-related toxicities have affected their clinical utility. The use of novel anti-CD3 binding domains with more favorable properties could aid in the creation of T-BsAbs with improved therapeutic windows. Using a sequence-based discovery platform, we identified new anti-CD3 antibodies from humanized rats that bind to multiple epitopes and elicit varying levels of T-cell activation. In T-BsAb format, 12 different anti-CD3 arms induce equivalent levels of tumor cell lysis by primary T-cells, but potency varies by a thousand-fold. Our lead CD3-targeting arm stimulates very low levels of cytokine release, but drives robust tumor antigen-specific killing in vitro and in a mouse xenograft model. This new CD3-targeting antibody underpins a next-generation T-BsAb platform in which potent cytotoxicity is uncoupled from high levels of cytokine release, which may lead to a wider therapeutic window in the clinic.


Subject(s)
Antibodies, Bispecific/metabolism , Antibodies, Monoclonal/metabolism , CD3 Complex/immunology , Neoplasms/therapy , T-Lymphocytes/immunology , Animals , Animals, Inbred Strains , Antigens, Neoplasm/immunology , Cytokines/metabolism , Cytotoxicity, Immunologic , Female , Humans , Jurkat Cells , Lymphocyte Activation , Mice , Neoplasms/immunology , Rats , Xenograft Model Antitumor Assays
8.
Front Immunol ; 9: 889, 2018.
Article in English | MEDLINE | ID: mdl-29740455

ABSTRACT

We created a novel transgenic rat that expresses human antibodies comprising a diverse repertoire of heavy chains with a single common rearranged kappa light chain (IgKV3-15-JK1). This fixed light chain animal, called OmniFlic, presents a unique system for human therapeutic antibody discovery and a model to study heavy chain repertoire diversity in the context of a constant light chain. The purpose of this study was to analyze heavy chain variable gene usage, clonotype diversity, and to describe the sequence characteristics of antigen-specific monoclonal antibodies (mAbs) isolated from immunized OmniFlic animals. Using next-generation sequencing antibody repertoire analysis, we measured heavy chain variable gene usage and the diversity of clonotypes present in the lymph node germinal centers of 75 OmniFlic rats immunized with 9 different protein antigens. Furthermore, we expressed 2,560 unique heavy chain sequences sampled from a diverse set of clonotypes as fixed light chain antibody proteins and measured their binding to antigen by ELISA. Finally, we measured patterns and overall levels of somatic hypermutation in the full B-cell repertoire and in the 2,560 mAbs tested for binding. The results demonstrate that OmniFlic animals produce an abundance of antigen-specific antibodies with heavy chain clonotype diversity that is similar to what has been described with unrestricted light chain use in mammals. In addition, we show that sequence-based discovery is a highly effective and efficient way to identify a large number of diverse monoclonal antibodies to a protein target of interest.


Subject(s)
Antibodies, Monoclonal/immunology , Drug Discovery/methods , Genes, Immunoglobulin Heavy Chain/genetics , Genes, Immunoglobulin Light Chain/genetics , Immunoglobulin kappa-Chains/immunology , Animals , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/therapeutic use , Antigens/administration & dosage , Antigens/immunology , B-Lymphocytes/immunology , Germinal Center/cytology , Germinal Center/immunology , High-Throughput Nucleotide Sequencing , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin kappa-Chains/genetics , Models, Animal , Rats , Rats, Sprague-Dawley , Rats, Transgenic
9.
Front Immunol ; 9: 3037, 2018.
Article in English | MEDLINE | ID: mdl-30666250

ABSTRACT

Heavy chain-only antibodies (HCAbs) do not associate with light chains and their VH regions are functional as single domains, forming the smallest active antibody fragment. These VH regions are ideal building blocks for a variety of antibody-based biologics because they tolerate fusion to other molecules and may also be attached in series to construct multispecific antibodies without the need for protein engineering to ensure proper heavy and light chain pairing. Production of human HCAbs has been impeded by the fact that natural human VH regions require light chain association and display poor biophysical characteristics when expressed in the absence of light chains. Here, we present an innovative platform for the rapid development of diverse sets of human HCAbs that have been selected in vivo. Our unique approach combines antibody repertoire analysis with immunization of transgenic rats, called UniRats, that produce chimeric HCAbs with fully human VH domains in response to an antigen challenge. UniRats express HCAbs from large transgenic loci representing the entire productive human heavy chain V(D)J repertoire, mount robust immune responses to a wide array of antigens, exhibit diverse V gene usage and generate large panels of stable, high affinity, antigen-specific molecules.


Subject(s)
Antibodies/chemistry , Antibodies/immunology , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Protein Engineering/methods , Animals , Antibody Affinity , Antigens/immunology , B-Lymphocytes/immunology , CHO Cells , Cricetulus , Crystallography , Flow Cytometry , Genetic Loci , High-Throughput Nucleotide Sequencing , Humans , Immunization , Immunoglobulin Light Chains/genetics , Immunoglobulin Variable Region/genetics , Immunoglobulin Variable Region/immunology , Protein Structure, Secondary , Rats , Rats, Transgenic , Single-Domain Antibodies/chemistry
10.
Am J Physiol Lung Cell Mol Physiol ; 312(5): L710-L721, 2017 05 01.
Article in English | MEDLINE | ID: mdl-28235950

ABSTRACT

Over past years, a critical role for the immune system and, in particular, for mast cells in the pathogenesis of pulmonary hypertension (PH) has emerged. However, the way in which mast cells promote PH is still poorly understood. Here, we investigated the mechanisms by which mast cells may contribute to PH, specifically focusing on the interaction between the innate and adaptive immune response and the role of B cells and autoimmunity. Experiments were performed in Sprague-Dawley rats and B cell-deficient JH-KO rats in the monocrotaline, Sugen/hypoxia, and the aortic banding model of PH. Hemodynamics, cell infiltration, IL-6 expression, and vascular remodeling were analyzed. Gene array analyses revealed constituents of immunoglobulins as most prominently regulated mast cell-dependent genes in the lung in experimental PH. IL-6 was shown to link mast cells to B cells, as 1) IL-6 was upregulated and colocalized with mast cells and was reduced by mast-cell stabilizers and 2) IL-6 or mast cell blockade reduced B cells in lungs of monocrotaline-treated rats. A functional role for B cells in PH was demonstrated in that either blocking B cells by an anti-CD20 antibody or B-cell deficiency in JH-KO rats attenuated right ventricular systolic pressure and vascular remodeling in experimental PH. We here identify a mast cell-B cell axis driven by IL-6 as a critical immune pathway in the pathophysiology of PH. Our results provide novel insights into the role of the immune system in PH, which may be therapeutically exploited by targeted immunotherapy.


Subject(s)
B-Lymphocytes/metabolism , Hypertension, Pulmonary/physiopathology , Lung/blood supply , Lung/physiopathology , Mast Cells/metabolism , Vascular Remodeling , Animals , Autoantibodies/metabolism , Blood Pressure , Heart Ventricles/pathology , Heart Ventricles/physiopathology , Interleukin-6/metabolism , Male , Models, Biological , Oligonucleotide Array Sequence Analysis , Rats, Sprague-Dawley , Systole , Time Factors
11.
Transplantation ; 101(8): 1770-1776, 2017 08.
Article in English | MEDLINE | ID: mdl-28230642

ABSTRACT

Techniques to obtain large quantities of antigen-specific monoclonal antibodies (mAbs) were first established in the 1970s when Georges Köhler and César Milstein immortalized antibody-producing mouse B-lymphocytes by fusion with myeloma cells (http://www.whatisbiotechnology.org/exhibitions/milstein). Combined with the expression of human antibodies in transgenic animals, this technique allowed upon immunization the generation of highly specific fully human mAbs for therapeutic applications. Apart from being extremely beneficial, mAbs are a huge success commercially. However, despite cell fusion generating many useful mAbs questions have been asked about which types of cells are prone to fuse and whether other methods may identify a wider range of binders. The discovery that expression libraries, using Escherichia coli or yeast, produced different specificities was intriguing and more recently Next-Generation Sequencing has identified wide-ranging usage with highly diverse and unique repertoires. Another strategy is the combination of flow cytometry sorting of antigen-binding B lymphocytes and single-cell reverse transcription polymerase chain reaction followed by reexpression, which has identified many high-affinity mAbs.


Subject(s)
Antibodies, Monoclonal/biosynthesis , B-Lymphocytes/immunology , Cell Fusion/methods , Immunization/methods , Animals , Animals, Genetically Modified , Flow Cytometry , Humans
12.
BMC Biotechnol ; 17(1): 3, 2017 01 09.
Article in English | MEDLINE | ID: mdl-28081707

ABSTRACT

BACKGROUND: There is an ever-increasing need of monoclonal antibodies (mAbs) for biomedical applications and fully human binders are particularly desirable due to their reduced immunogenicity in patients. We have applied a strategy for the isolation of antigen-specific B cells using tetramerized proteins and single-cell sorting followed by reconstruction of human mAbs by RT-PCR and expression cloning. RESULTS: This strategy, using human peripheral blood B cells, enabled the production of low affinity human mAbs against major histocompatibility complex molecules loaded with peptides (pMHC). We then implemented this technology using human immunoglobulin transgenic rats, which after immunization with an antigen of interest express high affinity-matured antibodies with human idiotypes. Using rapid immunization, followed by tetramer-based B-cell sorting and expression cloning, we generated several fully humanized mAbs with strong affinities, which could discriminate between highly homologous proteins (eg. different pMHC complexes). CONCLUSIONS: Therefore, we describe a versatile and more effective approach as compared to hybridoma generation or phage or yeast display technologies for the generation of highly specific and discriminative fully human mAbs that could be useful both for basic research and immunotherapeutic purposes.


Subject(s)
Antibodies, Monoclonal/genetics , Antibodies, Monoclonal/immunology , B-Lymphocytes/immunology , Cloning, Molecular/methods , Immunoglobulin G/immunology , Protein Engineering/methods , Animals , Antibodies, Monoclonal/biosynthesis , Cell Separation , Humans , Immunoglobulin G/genetics , Polymerase Chain Reaction , Rats
13.
Arch Immunol Ther Exp (Warsz) ; 63(2): 101-8, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25467949

ABSTRACT

Fully human antibodies from transgenic animals account for an increasing number of new therapeutics. After immunization, diverse human monoclonal antibodies of high affinity can be obtained from transgenic rodents, while large animals, such as transchromosomic cattle, have produced respectable amounts of specific human immunoglobulin (Ig) in serum. Several strategies to derive animals expressing human antibody repertoires have been successful. In rodents, gene loci on bacterial artificial chromosomes or yeast artificial chromosomes were integrated by oocyte microinjection or transfection of embryonic stem (ES) cells, while ruminants were derived from manipulated fibroblasts with integrated human chromosome fragments or human artificial chromosomes. In all strains, the endogenous Ig loci have been silenced by gene targeting, either in ES or fibroblast cells, or by zinc finger technology via DNA microinjection; this was essential for optimal production. However, comparisons showed that fully human antibodies were not as efficiently produced as wild-type Ig. This suboptimal performance, with respect to immune response and antibody yield, was attributed to imperfect interaction of the human constant region with endogenous signaling components such as the Igα/ß in mouse, rat or cattle. Significant improvements were obtained when the human V-region genes were linked to the endogenous CH-region, either on large constructs or, separately, by site-specific integration, which could also silence the endogenous Ig locus by gene replacement or inversion. In animals with knocked-out endogenous Ig loci and integrated large IgH loci, containing many human Vs, all D and all J segments linked to endogenous C genes, highly diverse human antibody production similar to normal animals was obtained.


Subject(s)
Antibodies, Monoclonal, Humanized/metabolism , Antibodies, Monoclonal/metabolism , Fibroblasts/physiology , Immunotherapy , Oocytes/physiology , Animals , Animals, Genetically Modified , Antibodies, Monoclonal/genetics , Antibodies, Monoclonal, Humanized/genetics , Cattle , Humans , Immunoglobulin Constant Regions/genetics , Mice , Microinjections , Protein Engineering , Rats , Species Specificity
14.
Genome Res ; 24(8): 1371-83, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24989021

ABSTRACT

The generation of genetically modified animals is important for both research and commercial purposes. The rat is an important model organism that until recently lacked efficient genetic engineering tools. Sequence-specific nucleases, such as ZFNs, TALE nucleases, and CRISPR/Cas9 have allowed the creation of rat knockout models. Genetic engineering by homology-directed repair (HDR) is utilized to create animals expressing transgenes in a controlled way and to introduce precise genetic modifications. We applied TALE nucleases and donor DNA microinjection into zygotes to generate HDR-modified rats with large new sequences introduced into three different loci with high efficiency (0.62%-5.13% of microinjected zygotes). Two of these loci (Rosa26 and Hprt1) are known to allow robust and reproducible transgene expression and were targeted for integration of a GFP expression cassette driven by the CAG promoter. GFP-expressing embryos and four Rosa26 GFP rat lines analyzed showed strong and widespread GFP expression in most cells of all analyzed tissues. The third targeted locus was Ighm, where we performed successful exon exchange of rat exon 2 for the human one. At all three loci we observed HDR only when using linear and not circular donor DNA. Mild hypothermic (30°C) culture of zygotes after microinjection increased HDR efficiency for some loci. Our study demonstrates that TALE nuclease and donor DNA microinjection into rat zygotes results in efficient and reproducible targeted donor integration by HDR. This allowed creation of genetically modified rats in a work-, cost-, and time-effective manner.


Subject(s)
Gene Targeting , Genetic Engineering , Animals , Base Sequence , Cells, Cultured , DNA Restriction Enzymes/biosynthesis , DNA Restriction Enzymes/genetics , Female , Hypoxanthine Phosphoribosyltransferase/genetics , Male , Microinjections , Rats, Sprague-Dawley , Rats, Transgenic , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinational DNA Repair , Zygote
15.
J Immunol Methods ; 400-401: 78-86, 2013 Dec 31.
Article in English | MEDLINE | ID: mdl-24184135

ABSTRACT

Expression of human antibody repertoires in transgenic animals has been accomplished by introducing large human Ig loci into mice and, more recently, a chimeric IgH locus into rats. With human VH, D and JH genes linked to the rat C-region antibody expression was significantly increased, similar to wild-type levels not found with fully human constructs. Here we compare four rat-lines containing the same human VH-region (comprising 22 VHs, all Ds and all JHs in natural configuration) but linked to different rat CH-genes and regulatory sequences. The endogenous IgH locus was silenced by zinc-finger nucleases. After breeding, all lines produced exclusively chimeric human H-chain with near normal IgM levels. However, in two lines poor IgG expression and inefficient immune responses were observed, implying that high expression, class-switching and hypermutation are linked to optimal enhancer function provided by the large regulatory region at the 3' end of the IgH locus. Furthermore, exclusion of Cδ and its downstream interval region may assist recombination. Highly diverse IgG and immune responses similar to normal rats were identified in two strains carrying diverse and differently spaced C-genes.


Subject(s)
Antibody Diversity/genetics , Genes, Immunoglobulin Heavy Chain/genetics , Genetic Loci , Immunoglobulin Fc Fragments/metabolism , Animals , Genes, Immunoglobulin Light Chain/genetics , Humans , Immunoglobulin Fc Fragments/genetics , Immunoglobulin Variable Region/genetics , Rats , Rats, Inbred Strains , Rats, Transgenic , Recombinant Fusion Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , VDJ Exons/genetics
16.
J Immunol ; 190(4): 1481-90, 2013 Feb 15.
Article in English | MEDLINE | ID: mdl-23303672

ABSTRACT

Mice transgenic for human Ig loci are an invaluable resource for the production of human Abs. However, such mice often do not yield human mAbs as effectively as conventional mice yield mouse mAbs. Suboptimal efficacy in delivery of human Abs might reflect imperfect interaction between the human membrane IgH chains and the mouse cellular signaling machinery. To obviate this problem, in this study we generated a humanized rat strain (OmniRat) carrying a chimeric human/rat IgH locus (comprising 22 human V(H)s, all human D and J(H) segments in natural configuration linked to the rat C(H) locus) together with fully human IgL loci (12 Vκs linked to Jκ-Cκ and 16 Vλs linked to Jλ-Cλ). The endogenous Ig loci were silenced using designer zinc finger nucleases. Breeding to homozygosity resulted in a novel transgenic rat line exclusively producing chimeric Abs with human idiotypes. B cell recovery was indistinguishable from wild-type animals, and human V(D)J transcripts were highly diverse. Following immunization, the OmniRat strain performed as efficiently as did normal rats in yielding high-affinity serum IgG. mAbs, comprising fully human variable regions with subnanomolar Ag affinity and carrying extensive somatic mutations, are readily obtainable, similarly to conventional mAbs from normal rats.


Subject(s)
Binding Sites, Antibody , IgG Deficiency/genetics , IgG Deficiency/immunology , Immunoglobulin Constant Regions/genetics , Immunoglobulin G/biosynthesis , Immunoglobulin Heavy Chains/metabolism , Immunoglobulin kappa-Chains/genetics , Immunoglobulin lambda-Chains/genetics , Animals , Binding Sites, Antibody/genetics , Chromosomes, Artificial, Bacterial/genetics , Chromosomes, Artificial, Yeast/genetics , Genes, Overlapping/genetics , Germ Cells/immunology , Germ Cells/metabolism , Humans , Immunoglobulin G/genetics , Immunoglobulin G/metabolism , Immunoglobulin Heavy Chains/genetics , Mice , Mice, Transgenic , Rats , Rats, Transgenic
17.
Int Immunol ; 23(10): 625-36, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21930595

ABSTRACT

The rat is an important biomedical experimental model that benefited from the recent development of new transgenic and knockout techniques. With the goal to optimize rat mAb production and to analyze the impact of Bcl-2 on B-cell development, we generated bcl-2 transgenic rats. Transgenic rats showed Bcl-2 over-expression in B cells, increased B cell numbers in lymphoid organs, elevated production of immunoglobulins (Igs) and prolonged B-cell survival in vitro. Transgenic rats remained healthy, reproduced normally and did not develop autoimmunity. Fusions with bcl-2 transgenic splenocytes did not result in increased hybridoma generation. A comparison of on- and off-rates of 39 mAbs generated with bcl-2 transgenic and wild-type animals revealed no significant differences. Over-expression of Bcl-2 in hybridomas did not change cell proliferation but resulted in increased Ig production. Bcl-2 transgenic rats will be a useful tool for the generation of rat mAbs, the analysis of B cells in different pathophysiological models, such as autoimmunity, cancer or organ transplantation, and the study of rat B-cell biology.


Subject(s)
B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Hybridomas/immunology , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Animals , B-Lymphocytes/cytology , Immunoglobulins/biosynthesis , Immunoglobulins/immunology , Rats , Rats, Sprague-Dawley , Rats, Transgenic
18.
Eur J Immunol ; 40(10): 2932-41, 2010 Oct.
Article in English | MEDLINE | ID: mdl-21038471

ABSTRACT

The rat is a species frequently used in immunological studies but, until now, there were no models with introduced gene-specific mutations. In a recent study, we described for the first time the generation of novel rat lines with targeted mutations using zinc-finger nucleases. In this study, we compare immune development in two Ig heavy-chain KO lines; one with truncated Cµ and a new line with removed JH segments. Rats homozygous for IgM mutation generate truncated Cµ mRNA with a de novo stop codon and no Cγ mRNA. JH-deletion rats showed undetectable mRNA for all H-chain transcripts. No serum IgM, IgG, IgA and IgE were detected in these rat lines. In both lines, lymphoid B-cell numbers were reduced >95% versus WT animals. In rats homozygous for IgM mutation, no Ab-mediated hyperacute allograft rejection was encountered. Similarities in B-cell differentiation seen in Ig KO rats and ES cell-derived Ig KO mice are discussed. These Ig and B-cell-deficient rats obtained using zinc-finger nucleases-technology should be useful as biomedical research models and a powerful platform for transgenic animals expressing a human Ab repertoire.


Subject(s)
B-Lymphocytes/immunology , Heart Transplantation/immunology , Immunoglobulin Constant Regions/immunology , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Joining Region/immunology , Amino Acid Sequence , Animals , Animals, Genetically Modified , B-Lymphocytes/cytology , Cell Differentiation/immunology , Embryonic Stem Cells/immunology , Graft Survival/immunology , Immunoglobulin Constant Regions/genetics , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Isotypes/blood , Immunoglobulin Joining Region/genetics , Lymphoid Tissue/immunology , Molecular Sequence Data , RNA/chemistry , RNA/genetics , Rats , Rats, Inbred Lew , Rats, Mutant Strains , Rats, Sprague-Dawley , Reverse Transcriptase Polymerase Chain Reaction , Specific Pathogen-Free Organisms , Zinc Fingers/genetics
19.
Clin Vaccine Immunol ; 17(6): 930-6, 2010 Jun.
Article in English | MEDLINE | ID: mdl-20410329

ABSTRACT

New Zealand White (NZW) rabbits were immunized with several different nontoxic botulinum neurotoxin serotype B (BoNT/B) preparations in an effort to optimize the production of a rapid and highly potent, effective neutralizing antibody response. The immunogens included a recombinant heavy chain (rHc) protein produced in Escherichia coli, a commercially available formaldehyde-inactivated toxoid, and an alkylated toxoid produced by urea-iodoacetamide inactivation of the purified active toxin. All three immunogens elicited an antibody response to BoNT/B, detected by enzyme-linked immunosorbent assay (ELISA) and by toxin neutralization assay, by the use of two distinct mouse toxin challenge models. The induction period and the ultimate potency of the observed immune response varied for each immunogen, and the ELISA titer was not reliably predictive of the potency of toxin neutralization. The kinetics of the BoNT/B-specific binding immune response were nearly identical for the formaldehyde toxoid and alkylated toxoid immunogens, but immunization with the alkylated toxoid generated an approximately 10-fold higher neutralization potency that endured throughout the study, and after just 49 days, each milliliter of serum was capable of neutralizing 10(7) 50% lethal doses of the toxin. Overall, the immunization of rabbits with alkylated BoNT/B toxoid appears to have induced a neutralizing immune response more rapid and more potent than the responses generated by vaccination with formaldehyde toxoid or rHc preparations.


Subject(s)
Antibodies, Neutralizing/blood , Botulinum Toxins/immunology , Iodoacetamide/pharmacology , Toxoids/administration & dosage , Alkylating Agents/pharmacology , Animals , Antibodies, Bacterial/blood , Antibodies, Bacterial/immunology , Antibodies, Neutralizing/immunology , Botulinum Toxins/toxicity , Botulinum Toxins, Type A , Enzyme-Linked Immunosorbent Assay , Female , Mice , Neutralization Tests , Rabbits , Time Factors , Toxoids/chemistry , Toxoids/immunology
20.
Methods Mol Biol ; 597: 211-25, 2010.
Article in English | MEDLINE | ID: mdl-20013236

ABSTRACT

The genetic dissection of physiological and pathological traits in laboratory model organisms is accelerated by the ability to engineer loss-of-function mutations at investigator-specified loci. This chapter describes the use of zinc-finger nucleases (ZFNs) for the targeted disruption of endogenous rat genes directly in the embryo. ZFNs can specifically disrupt target genes in cultured rat cells and in embryos from inbred and outbred strains, leading to permanently genetically modified animals. This technology allows for the rapid, targeted modification of the rat genome.


Subject(s)
Animals, Genetically Modified/genetics , Deoxyribonucleases, Type II Site-Specific/genetics , Genes , Rats/genetics , Zinc Fingers , Animals , Cells, Cultured , Deoxyribonucleases, Type II Site-Specific/analysis , Deoxyribonucleases, Type II Site-Specific/metabolism , Embryo Transfer , Female , Gene Knockout Techniques/methods , Genome , Male , Protein Engineering/methods , RNA, Messenger/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...