Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Proc Natl Acad Sci U S A ; 119(32): e2200879119, 2022 08 09.
Article in English | MEDLINE | ID: mdl-35925889

ABSTRACT

The value of anti-CTLA-4 antibodies in cancer therapy is well established. However, the broad application of currently available anti-CTLA-4 therapeutic antibodies is hampered by their narrow therapeutic index. It is therefore challenging and attractive to develop the next generation of anti-CTLA-4 therapeutics with improved safety and efficacy. To this end, we generated fully human heavy chain-only antibodies (HCAbs) against CTLA-4. The hIgG1 Fc domain of the top candidate, HCAb 4003-1, was further engineered to enhance its regulatory T (Treg) cell depletion effect and to decrease its half-life, resulting in HCAb 4003-2. We tested these HCAbs in in vitro and in vivo experiments in comparison with ipilimumab and other anti-CTLA4 antibodies. The results show that human HCAb 4003-2 binds human CTLA-4 with high affinity and potently blocks the binding of B7-1 (CD80) and B7-2 (CD86) to CTLA-4. The results also show efficient tumor penetration. HCAb 4003-2 exhibits enhanced antibody-dependent cellular cytotoxicity function, lower serum exposure, and more potent anti-tumor activity than ipilimumab in murine tumor models, which is partly driven by a substantial depletion of intratumoral Tregs. Importantly, the enhanced efficacy combined with the shorter serum half-life and less systemic drug exposure in vivo potentially provides an improved therapeutic window in cynomolgus monkeys and preliminary clinical applications. With its augmented efficacy via Treg depletion and improved safety profile, HCAb 4003-2 is a promising candidate for the development of next generation anti-CTLA-4 therapy.


Subject(s)
Immunoglobulin Heavy Chains , Immunotherapy , Neoplasms , T-Lymphocytes, Regulatory , Animals , Antibody-Dependent Cell Cytotoxicity , CTLA-4 Antigen/immunology , Humans , Immunoglobulin Heavy Chains/pharmacology , Ipilimumab/pharmacology , Mice , Neoplasms/pathology , Neoplasms/therapy
2.
Biochem Biophys Res Commun ; 565: 1-7, 2021 08 06.
Article in English | MEDLINE | ID: mdl-34077827

ABSTRACT

Streptococcus pyogenes causes a wide range of human infections. Currently, antibiotics are the main treatment for S. pyogenes infection, but serious anti-microbial resistance requires alternative treatment options. To develop a novel strategy for treatment, we physicochemically characterized SPs0871, a putative maltose/maltodextrin-binding protein that is thought to have important roles in the pathogenesis of invasive streptococci. We obtained a variable domain of heavy chain of heavy-chain antibody, the smallest unit of an antibody, which specifically binds to SPs0871. Although the VHH completely inhibited the binding of maltodextrins to SPs0871, the inhibition did not lead to growth suppression of the bacteria. Our results provide important insights for development of VHH as an anti-streptococcal therapeutic.


Subject(s)
Anti-Bacterial Agents/pharmacology , Bacterial Proteins/antagonists & inhibitors , Immunoglobulin Heavy Chains/pharmacology , Polysaccharides/antagonists & inhibitors , Streptococcus pyogenes/drug effects , Anti-Bacterial Agents/chemistry , Bacterial Proteins/chemistry , Immunoglobulin Heavy Chains/chemistry , Microbial Sensitivity Tests , Polysaccharides/chemistry , Streptococcus pyogenes/chemistry
3.
Protein Expr Purif ; 170: 105596, 2020 06.
Article in English | MEDLINE | ID: mdl-32036001

ABSTRACT

Antibodies that block interaction of immune checkpoint receptors with its ligands have revolutionized the treatment of several cancers. Despite the success of this approach, the high cost has been restricted the use of this class of drugs. In this context, the development of biosimilar can be an important strategy for reducing prices and expanding access after patent has been dropped. Here, we evaluated the use of HEK293 cells for transient expression of an immune checkpoint-blocking antibody as a first step for biosimilar development. Antibody light and heavy chain genes were cloned into pCI-neo vector and transiently expressed in HEK293 cells. The culture supernatant was then subjected to protein A affinity chromatography, which allowed to obtain the antibody with high homogeneity. For physicochemical comparability, biosimilar antibody and reference drug were analyzed by SDS-PAGE, isoelectric focusing, circular dichroism and fluorescence spectroscopy. The results indicated that the both antibodies have a high degree of structural similarity. Lastly, the biosimilar antibody binding capacity to target receptor was shown to be similar to reference product in ELISA and flow cytometry assays. These data demonstrate that the HEK293 system can be used as an important tool for candidate selection and early development of biosimilar antibodies.


Subject(s)
Antibodies, Monoclonal/pharmacology , Biosimilar Pharmaceuticals/pharmacology , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/genetics , Immunoglobulin Heavy Chains/pharmacology , Immunoglobulin Light Chains/pharmacology , Antibodies, Monoclonal/biosynthesis , Antibody Affinity , Antibody Specificity , Biosimilar Pharmaceuticals/metabolism , Chromatography, Affinity , Genetic Vectors/chemistry , Genetic Vectors/metabolism , HEK293 Cells , Humans , Immune Checkpoint Inhibitors/immunology , Immune Checkpoint Proteins/immunology , Immunoglobulin Heavy Chains/biosynthesis , Immunoglobulin Light Chains/biosynthesis , Isoelectric Focusing
4.
Bull Exp Biol Med ; 161(1): 92-5, 2016 May.
Article in English | MEDLINE | ID: mdl-27265131

ABSTRACT

Major histocompatibility complex class II (MHC II) plays an important role not only in the adaptive immune responses to foreign pathogens, but also in the development of some autoimmune diseases. Non-classical MHC, HLA-DM is directly involved in MHC II loading with the peptide. To study this process, we synthesized recombinant proteins HLA-DR1 and HLA-DM. α/ß-Chains of DR1 heterodimer contained C-terminal leucine domains of the fos and jun factors, respectively. Each DM chain contained constant fragment of human antibody heavy chain fused via a long linker domain. In addition, DM α-chain carried N165D substitution suppressing potential glycosylation at this site. We observed significant acceleration of DR1 peptide loading with influenza HA306-318 hemagglutinin in the presence of DM, which indicates functionality of recombinant DR1-DM protein couple. Our results can be used to study the presentation of other viral and self-antigens and can become the basis for the development of new drug modeling.


Subject(s)
HLA-D Antigens/pharmacology , HLA-DR1 Antigen/physiology , Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoglobulin Constant Regions/pharmacology , Immunoglobulin Heavy Chains/pharmacology , Peptide Fragments/immunology , Recombinant Fusion Proteins/pharmacology , Adaptive Immunity , Animals , Antigen Presentation/drug effects , Autoimmune Diseases/immunology , Autoimmunity , Drosophila melanogaster , HEK293 Cells , HeLa Cells , Humans , Protein Binding
5.
Int J Mol Sci ; 15(6): 9481-96, 2014 May 28.
Article in English | MEDLINE | ID: mdl-24879522

ABSTRACT

OBJECTIVE: To construct an immune alpaca phage display library, in order to obtain a single domain anti-BAFF (B cell-activating factor) antibody. METHODS: Using phage display technology, we constructed an immune alpaca phage display library, selected anti-BAFF single domain antibodies (sdAbs), cloned three anti-BAFF single-domain antibody genes into expression vector pSJF2, and expressed them efficiently in Escherichia coli. The affinity of different anti-BAFF sdAbs were measured by Bio layer interferometry. The in vitro biological function of three sdAbs was investigated by cell counting kit-8 (CCK-8) assay and a competitive enzyme-linked immunosorbent assay (ELISA). RESULTS: We obtained three anti-BAFF single domain antibodies (anti-BAFF64, anti-BAFF52 and anti-BAFFG3), which were produced in high yield in Escherichia coli and inhibited tumor cell proliferation in vitro. CONCLUSION: The selected anti-BAFF antibodies could be candidates for B-cell lymphoma therapies.


Subject(s)
B-Cell Activating Factor/immunology , Lymphoma, B-Cell/drug therapy , Peptide Library , Single-Domain Antibodies/immunology , Single-Domain Antibodies/pharmacology , Amino Acid Sequence , Animals , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Camelids, New World , Cell Line, Tumor , Cell Proliferation/drug effects , Cloning, Molecular , Escherichia coli/genetics , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/genetics , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/pharmacology , Lymphoma, B-Cell/immunology , Molecular Sequence Data , Sequence Alignment , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/genetics
6.
J Biol Chem ; 287(18): 14912-22, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22334655

ABSTRACT

Complementarity-determining regions (CDRs) from monoclonal antibodies tested as synthetic peptides display anti-infective and antitumor activities, independent of the specificity of the native antibody. Previously, we have shown that the synthetic peptide C7H2, based on the heavy chain CDR 2 from monoclonal antibody C7, a mAb directed to a mannoprotein of Candida albicans, significantly reduced B16F10 melanoma growth and lung colony formation by triggering tumor apoptosis. The mechanism, however, by which C7H2 induced apoptosis in tumor cells remained unknown. Here, we demonstrate that C7H2 interacts with components of the tumor cells cytoskeleton, being rapidly internalized after binding to the tumor cell surface. Mass spectrometry analysis and in vitro validation revealed that ß-actin is the receptor of C7H2 in the tumor cells. C7H2 induces ß-actin polymerization and F-actin stabilization, linked with abundant generation of superoxide anions and apoptosis. Major phenotypes following peptide binding were chromatin condensation, DNA fragmentation, annexin V binding, lamin disruption, caspase 8 and 3 activation, and organelle alterations. Finally, we evaluated the cytotoxic efficacy of C7H2 in a panel of human tumor cell lines. All tumor cell lines studied were equally susceptible to C7H2 in vitro. The C7H2 amide without further derivatization significantly reduced lung metastasis of mice endovenously challenged with B16F10-Nex2 melanoma cells. No significant cytotoxicity was observed toward nontumorigenic cell lines on short incubation in vitro or in naïve mice injected with a high dose of the peptide. We believe that C7H2 is a promising peptide to be developed as an anticancer drug.


Subject(s)
Actins/immunology , Antibodies, Monoclonal, Murine-Derived/pharmacology , Antibodies, Neoplasm/pharmacology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Immunoglobulin Heavy Chains/pharmacology , Immunoglobulin Variable Region/pharmacology , Melanoma/prevention & control , Neoplasm Proteins/immunology , Animals , Antibodies, Monoclonal, Murine-Derived/immunology , Antineoplastic Agents/immunology , Candida albicans/immunology , Caspase 3/immunology , Caspase 8/immunology , Cell Line, Tumor , DNA Fragmentation/drug effects , DNA, Neoplasm/immunology , Fungal Proteins/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Variable Region/immunology , Male , Melanoma/immunology , Melanoma/pathology , Membrane Glycoproteins/immunology , Mice , Neoplasm Metastasis
7.
Blood ; 118(3): 757-65, 2011 Jul 21.
Article in English | MEDLINE | ID: mdl-21576702

ABSTRACT

Neutralizing the interaction of the platelet receptor gpIb with VWF is an attractive strategy to treat and prevent thrombotic complications. ALX-0081 is a bivalent Nanobody which specifically targets the gpIb-binding site of VWF and interacts avidly with VWF. Nanobodies are therapeutic proteins derived from naturally occurring heavy-chain-only Abs and combine a small molecular size with a high inherent stability. ALX-0081 exerts potent activity in vitro and in vivo. Perfusion experiments with blood from patients with acute coronary syndrome on standard antithrombotics demonstrated complete inhibition of platelet adhesion after addition of ALX-0081, while in the absence of ALX-0081 residual adhesion was observed. In a baboon efficacy and safety model measuring acute thrombosis and surgical bleeding, ALX-0081 showed a superior therapeutic window compared with marketed antithrombotics. Pharmacokinetic and biodistribution experiments demonstrated target-mediated clearance of ALX-0081, which leads to a self-regulating disposition behavior. In conclusion, these preclinical data demonstrate that ALX-0081 combines a high efficacy with an improved safety profile compared with currently marketed antithrombotics. ALX-0081 has entered clinical development.


Subject(s)
Antibodies, Bispecific/pharmacokinetics , Fibrinolytic Agents/pharmacology , Immunoglobulin Heavy Chains/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Single-Chain Antibodies/pharmacokinetics , Thrombosis/drug therapy , Animals , Antibody Specificity , Binding Sites/immunology , Fibrinolytic Agents/immunology , Humans , In Vitro Techniques , Macaca fascicularis , Papio , Platelet Adhesiveness/drug effects , Platelet Adhesiveness/immunology , Platelet Glycoprotein GPIb-IX Complex/immunology , Platelet Glycoprotein GPIb-IX Complex/metabolism , Pulsatile Flow/physiology , Thrombosis/immunology , von Willebrand Factor/immunology , von Willebrand Factor/metabolism
8.
Viruses ; 12(10)2020 10 08.
Article in English | MEDLINE | ID: mdl-33049994

ABSTRACT

Broadly neutralizing monoclonal antibodies (bNAbs) against conserved domains in the influenza hemagglutinin are in clinical trials. Several next generation influenza vaccines designed to elicit such bNAbs are also in clinical development. One of the common features of the isolated bNAbs is the use of restricted IgVH repertoire. More than 80% of stem-targeting bNAbs express IgVH1-69, which may indicate genetic constraints on the evolution of such antibodies. In the current study, we evaluated a panel of influenza virus bNAbs in comparison with HIV-1 MAb 4E10 and anti-RSV MAb Palivizumab (approved for human use) for autoreactivity using 30 normal human tissues microarray and human protein (>9000) arrays. We found that several human bNAbs (CR6261, CR9114, and F2603) reacted with human tissues, especially with pituitary gland tissue. Importantly, protein array analysis identified high-affinity interaction of CR6261 with the autoantigen "Enhancer of mRNA decapping 3 homolog" (EDC3), which was not previously described. Moreover, EDC3 competed with hemagglutinin for binding to bNAb CR6261. These autoreactivity findings underscores the need for careful evaluation of such bNAbs for therapeutics and stem-based vaccines against influenza virus.


Subject(s)
Hemagglutinin Glycoproteins, Influenza Virus/immunology , Immunoglobulin Heavy Chains/pharmacology , Influenza Vaccines/adverse effects , Influenza Vaccines/immunology , Influenza, Human/prevention & control , Single-Chain Antibodies/pharmacology , Antibodies, Monoclonal/immunology , Antibodies, Neutralizing/immunology , Antibodies, Neutralizing/pharmacology , Antibodies, Viral/immunology , Antibodies, Viral/pharmacology , Autoantibodies/immunology , Broadly Neutralizing Antibodies/immunology , HIV Antibodies/immunology , HIV-1/immunology , Humans , Immunoglobulin Heavy Chains/immunology , Ribonucleoproteins, Small Nuclear/immunology , Single-Chain Antibodies/immunology
9.
J Leukoc Biol ; 107(6): 933-939, 2020 06.
Article in English | MEDLINE | ID: mdl-32040234

ABSTRACT

Systemic TNF neutralization can be used as a therapy for several autoimmune diseases. To evaluate the effects of cell type-restricted TNF blockade, we previously generated bispecific antibodies that can limit TNF secretion by myeloid cells (myeloid cell-specific TNF inhibitors or MYSTIs). In this study several such variable domain (VH) of a camelid heavy-chain only antibody-based TNF inhibitors were compared in relevant experimental models, both in vitro and in vivo. Pretreatment with MYSTI-2, containing the anti-F4/80 module, can restrict the release of human TNF (hTNF) from LPS-activated bone marrow-derived macrophage (BMDM) cultures of humanized TNF knock-in (mice; hTNFKI) more effectively than MYSTI-3, containing the anti-CD11b module. MYSTI-2 was also superior to MYSTI-3 in providing in vivo protection in acute toxicity model. Finally, MYSTI-2 was at least as effective as Infliximab in preventing collagen antibody-induced arthritis. This study demonstrates that a 33 kDa bispecific mini-antibody that specifically restricts TNF secretion by macrophages is efficient for amelioration of experimental arthritis.


Subject(s)
Antibodies, Monoclonal/pharmacology , Arthritis, Experimental/therapy , CD11b Antigen/antagonists & inhibitors , Calcium-Binding Proteins/antagonists & inhibitors , Immunoglobulin Heavy Chains/pharmacology , Myeloid Progenitor Cells/drug effects , Receptors, G-Protein-Coupled/antagonists & inhibitors , Tumor Necrosis Factor Inhibitors/pharmacology , Animals , Antirheumatic Agents/pharmacology , Arthritis, Experimental/genetics , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , CD11b Antigen/genetics , CD11b Antigen/immunology , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/immunology , Gene Expression , Humans , Infliximab/pharmacology , Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/immunology , Mice , Mice, Transgenic , Myeloid Progenitor Cells/immunology , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/immunology , Tumor Necrosis Factor-alpha/genetics , Tumor Necrosis Factor-alpha/immunology
10.
Mol Immunol ; 45(4): 881-6, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17889938

ABSTRACT

Active and passive immunotherapy targeted at the amyloid-beta (Abeta) peptide has been proposed as therapeutic approach against Alzheimer's disease (AD), and efforts towards the generation and application of antibody-based reagents that are capable of preventing and clearing amyloid aggregates are currently under active investigation. Previously, we selected and characterized a new anti-Abeta1-42 phage-displayed scFv antibody, designated clone b4.4, using a non-immune human scFv antibody library and demonstrated that a peptide based on the sequence of the Ig heavy chain (VH) complementarity-determining region (HCDR3) of this antibody fragment bound to Abeta1-42)and had neuroprotective potential against Abeta1-42 mediated neurotoxicity in rat hippocampal cultured neurons. In the present study, using novel computational methods and in vitro experiments we demonstrated that b4.4 binds to the central region of Abeta1-42. We also demonstrated that this scFv antibody binds to Abeta-derived diffusible ligands (ADDLs) and neutralizes the toxicity of both fibrillar and oligomeric forms of Abeta1-42 tested in vitro in SH-SY5Y cell cultures.


Subject(s)
Amyloid beta-Peptides/pharmacology , Complementarity Determining Regions/pharmacology , Immunoglobulin Fragments/pharmacology , Immunoglobulin Heavy Chains/pharmacology , Neuroprotective Agents/pharmacology , Oligopeptides/pharmacology , Peptide Fragments/pharmacology , Amyloid beta-Peptides/chemistry , Amyloid beta-Peptides/immunology , Cell Line, Tumor , Cell Survival/drug effects , Complementarity Determining Regions/chemistry , Epitope Mapping , Humans , Immunoglobulin Fragments/chemistry , Immunoglobulin Heavy Chains/chemistry , Models, Molecular , Neuroprotective Agents/chemistry , Oligopeptides/chemistry , Peptide Fragments/chemistry , Peptide Fragments/immunology , Protein Binding
11.
Sci Rep ; 8(1): 7450, 2018 05 10.
Article in English | MEDLINE | ID: mdl-29748553

ABSTRACT

Nucleolin arises as a relevant target for cancer therapy, as it is overexpressed at the surface of cancer and angiogenic endothelial cells thus enabling a dual cellular targeting strategy. Immunotherapeutic strategies, albeit of proven therapeutic relevance, have been scarcely explored against this target. Therefore, this work aimed at engineering an anti-nucleolin VHH-based antibody capable of triggering anticancer immune responses. Herein, anti-nucleolin VHHs have been generated upon grafting F3 peptide-derived nucleolin-binding sequences onto a VHH CDR1 or CDR3. One of these nucleolin-binding CDR3-grafted VHH was subsequently fused to a human IgG1 Fc region, enabling a significant antibody-dependent cell-mediated cytotoxicity (ADCC). The generated anti-nucleolin VHH revealed increased binding and antiproliferative effects against cancer cells, relative to the parental VHH, while the VHH-Fc counterpart presented increased cytotoxicity relative to the corresponding VHH. This VHH-Fc also triggered an ADCC effect, in the nanomolar range, against a nucleolin-overexpressing cancer cell line. This effect was evidenced by a 2 or 1.7-fold increase of cell death, in the presence of PBMCs, relative to the parental VHH-Fc or the VHH counterpart, respectively. Overall, these formats represent the first anti-nucleolin VHHs and the first anti-nucleolin antibody with ADCC activity that have been successfully developed.


Subject(s)
Antibody-Dependent Cell Cytotoxicity/drug effects , Antineoplastic Agents, Immunological/pharmacology , Immunoglobulin Heavy Chains/pharmacology , Neoplasms/drug therapy , Phosphoproteins/immunology , RNA-Binding Proteins/immunology , Single-Domain Antibodies/pharmacology , Antineoplastic Agents, Immunological/immunology , Cell Death/drug effects , Cell Line, Tumor , Humans , Immunoglobulin Fc Fragments/immunology , Immunoglobulin Fc Fragments/pharmacology , Immunoglobulin G/immunology , Immunoglobulin G/pharmacology , Immunoglobulin Heavy Chains/immunology , Neoplasms/immunology , Single-Domain Antibodies/immunology , Nucleolin
12.
Sci Rep ; 7(1): 7438, 2017 08 07.
Article in English | MEDLINE | ID: mdl-28785006

ABSTRACT

Antibody treatment is currently the only available countermeasure for botulism, a fatal illness caused by flaccid paralysis of muscles due to botulinum neurotoxin (BoNT) intoxication. Among the seven major serotypes of BoNT/A-G, BoNT/A poses the most serious threat to humans because of its high potency and long duration of action. Prior to entering neurons and blocking neurotransmitter release, BoNT/A recognizes motoneurons via a dual-receptor binding process in which it engages both the neuron surface polysialoganglioside (PSG) and synaptic vesicle glycoprotein 2 (SV2). Previously, we identified a potent neutralizing antitoxin against BoNT/A1 termed ciA-C2, derived from a camelid heavy-chain-only antibody (VHH). In this study, we demonstrate that ciA-C2 prevents BoNT/A1 intoxication by inhibiting its binding to neuronal receptor SV2. Furthermore, we determined the crystal structure of ciA-C2 in complex with the receptor-binding domain of BoNT/A1 (HCA1) at 1.68 Å resolution. The structure revealed that ciA-C2 partially occupies the SV2-binding site on HCA1, causing direct interference of HCA1 interaction with both the N-glycan and peptide-moiety of SV2. Interestingly, this neutralization mechanism is similar to that of a monoclonal antibody in clinical trials, despite that ciA-C2 is more than 10-times smaller. Taken together, these results enlighten our understanding of BoNT/A1 interactions with its neuronal receptor, and further demonstrate that inhibiting toxin binding to the host receptor is an efficient countermeasure strategy.


Subject(s)
Antibodies, Neutralizing/pharmacology , Botulinum Toxins, Type A/chemistry , Botulinum Toxins, Type A/metabolism , Camelidae/immunology , Nerve Tissue Proteins/metabolism , Animals , Antibodies, Neutralizing/chemistry , Binding Sites , Crystallography, X-Ray , Gangliosides/metabolism , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/pharmacology , Models, Molecular , Protein Binding , Protein Conformation , Rats , Single-Domain Antibodies/chemistry , Single-Domain Antibodies/pharmacology
13.
J Leukoc Biol ; 78(4): 1001-7, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16081594

ABSTRACT

Transfer of B6 T cells to major histocompatibility complex (MHC) class I disparate bm1 x B6 F1 mice leads to the development of hepatic graft-versus-host disease (GVHD) characterized by an active hepatitis with portal and lobular inflammation as well as bile duct inflammation and venulitis. The present studies determined the role of tumor necrosis factor (TNF) in hepatic GVHD. B6 responder cells were cultured with irradiated MHC class I disparate bm1 or syngeneic spleen cells (SpC) in the presence or absence of TNF receptor inhibitor [TNFR-immunoglobulin (Ig)]. Recipient bm1 x B6 F1 mice were irradiated (600 cGy) and reconstituted with 5 x 10(6) T cell-depleted B6 bone marrow cells and 1 x 10(7) B6 SpC. Mice were injected with an adenovirus encoding TNFR-Ig [TNF inhibitor-encoding adenovirus (Adv-TNFi)] or beta-galactosidase (Adv-betagal). Severity of liver GVHD was assessed by a composite histopathological score consisting of the sum of scores for venulitis, lobular hepatitis, and bile duct inflammation. Addition of TNFR-Ig reduced cell proliferation in mixed lymphocyte cultures using B6 responder SpC by 71% +/- 12.8% and interferon-gamma responses by 78% +/- 18%. GVHD-induced "wasting disease" was reduced in Adv-TNFi recipients [4.4%+/-5.2% weight loss (n=11)] compared with Adv-betagal recipients [16.1%+/-7.6% weight loss (n=11; P=0.0004)] 9 days post-transplant. Composite histopathological scores and individual venulitis scores were reduced with the addition of Adv-TNFi. Hepatic CD8+ T cells in the recipients of Adv-TNFi were reduced as compared with recipients of Adv-betagal. In conclusion, Adv-TNFi reduces MHC class I disparate alloproliferative responses and hepatic GVHD.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Graft vs Host Disease/immunology , Histocompatibility Antigens Class I/immunology , Liver/pathology , Tumor Necrosis Factor-alpha/physiology , Animals , CD8-Positive T-Lymphocytes/drug effects , Cells, Cultured , Female , Histocompatibility Antigens Class I/drug effects , Immunoglobulin Heavy Chains/pharmacology , Immunoglobulin gamma-Chains , Interferon-gamma/biosynthesis , Interferon-gamma/drug effects , Interferon-gamma/immunology , Liver/immunology , Male , Mice , Mice, Inbred C57BL , Receptors, Tumor Necrosis Factor , Recombinant Fusion Proteins/pharmacology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Tumor Necrosis Factor-alpha/pharmacology
14.
MAbs ; 8(4): 761-74, 2016.
Article in English | MEDLINE | ID: mdl-26963639

ABSTRACT

We previously described 4Dm2m, an exceptionally potent broadly neutralizing CD4-antibody fusion protein against HIV-1. It was generated by fusing the engineered single human CD4 domain mD1.22 to both the N and C termini of the human IgG1 heavy chain constant region and the engineered single human antibody domain m36.4, which targets the CD4-induced coreceptor binding site of the viral envelope glycoprotein, to the N terminus of the human antibody kappa light chain constant region via the (G4S)3 polypeptide linkers. However, therapeutic use of 4Dm2m was limited by its short in vivo half-life. Here, we show that a combination of three approaches have successfully increased the persistence of 4Dm2m in mice. First, to stabilize the scaffold, we enhanced heterodimerization between the heavy chain constant domain 1 (CH1) and kappa light chain constant domain (CK) by using structure-guided design and phage-display library technologies. Second, to address the possibility that long polypeptide linkers might render fusion proteins more susceptible to proteolysis, we shortened the (G4S)3 linkers or replaced them with the human IgG1 hinge sequence, which is naturally designed for both flexibility and stability. Third, we introduced two amino acid mutations into the crystallizable fragment (Fc) of the scaffold previously shown to increase antibody binding to the neonatal Fc receptor (FcRn) and prolong half-lives in vivo. Collectively, these approaches markedly increased the serum concentrations of 4Dm2m in mice while not affecting other properties of the fusion protein. The new 4Dm2m variants are promising candidates for clinical development to prevent or treat HIV-1 infection. To our knowledge, this is the first report on stabilized CH1-CK, which is potentially useful as a new heterodimerization scaffold for generation of bispecific and multispecific antibodies or proteins with a more favorable pharmacokinetic profile.


Subject(s)
AIDS Vaccines/pharmacokinetics , Antibodies, Monoclonal/pharmacokinetics , Antibodies, Neutralizing/pharmacology , AIDS Vaccines/chemistry , Animals , Antibodies, Monoclonal/chemistry , Antibodies, Neutralizing/chemistry , Broadly Neutralizing Antibodies , CD4 Antigens , HIV-1/immunology , Half-Life , Humans , Immunoglobulin Constant Regions/chemistry , Immunoglobulin Constant Regions/pharmacology , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/pharmacology , Immunoglobulin kappa-Chains/chemistry , Immunoglobulin kappa-Chains/pharmacology , Mice , Mice, Inbred C57BL , Protein Engineering/methods , Protein Stability , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/pharmacokinetics
15.
J Mol Biol ; 289(1): 41-55, 1999 May 28.
Article in English | MEDLINE | ID: mdl-10339404

ABSTRACT

A library of Fd fragment antibody binding proteins was created by random mutation of 15 nucleotides within the CDRIII region of the immunoglobulin heavy chain gene and displayed as Fd coat protein fusion constructs of M13 phage. The library was screened for those VHbinding sites that bound glucose-6-phosphate dehydrogenase (G6PD). One isolate (DH27bp) inhibited G6PD activity by 85 %. The DH27bpgene was re-engineered, placed in a eukaryotic expression vector having an isopropyl-beta-delta-thiogalactopyranoside (IPTG) inducible promoter, and transfected and then expressed in Chinese hamster V79 cells. G6PD activity was completely inhibited. Removal of IPTG reverted the cell to full G6PD activity. The intracellular dynamics of the G6PD/DH27bpcomplex showed that when the proteasomes of cells expressing DH27bpwere inhibited (N -acetyl-Leu-Leu-norleucinal or lactacystin) G6PD activity increased. Metabolic labelling of newly synthesized IPTG-induced proteins during/absence of proteasomal inhibitors showed that both G6PD and DH27bpare signaled for degradation when the intracellular complex is formed. Furthermore, semi-quantitative RT/PCR demonstrated that G6PD mRNA is upregulated over the time course of G6PD inactivation by DH27bpFd binding protein. These effects were not observed in those cells expressing a non-mutated Fd (UMHC) or in IPTG-treated non-transduced V79 cells. Our results demonstrate that an Fd-based intracellular binding protein can find and disable the function of a specific intracellular target and once the Fd expression is repressed the activity of intracellular targeted protein can revert to normal.


Subject(s)
Glucosephosphate Dehydrogenase/antagonists & inhibitors , Glucosephosphate Dehydrogenase/genetics , Immunoglobulin Constant Regions/pharmacology , Immunoglobulin Heavy Chains/pharmacology , Immunoglobulin Variable Region/pharmacology , Acetylcysteine/analogs & derivatives , Acetylcysteine/pharmacology , Animals , Cell Line , Cricetinae , Cricetulus , Cysteine Endopeptidases/metabolism , Cysteine Proteinase Inhibitors/pharmacology , Cytoplasm/enzymology , Gene Expression Regulation, Enzymologic , Glucosephosphate Dehydrogenase/immunology , Inovirus , Isopropyl Thiogalactoside/pharmacology , Kinetics , Leupeptins/pharmacology , Multienzyme Complexes/metabolism , Peptide Library , Proteasome Endopeptidase Complex , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/pharmacology , Reverse Transcriptase Polymerase Chain Reaction , Transfection
16.
J Leukoc Biol ; 61(2): 141-6, 1997 Feb.
Article in English | MEDLINE | ID: mdl-9021918

ABSTRACT

Two types of cytotoxic reactions have been observed using cells from the nurse shark: spontaneous cytotoxicity mediated by cells of the macrophage lineage and antibody-dependent killing carried out by a different effector cell population. Previous data showed that removal of phagocytic cells using iron particles abolished macrophage-mediated killing, but not antibody-dependent reactions. The current study used single cell assays and showed that the effector of antibody-driven reactions was the neutrophil. Surprisingly, the mechanism of killing was shown to be phagocytosis mediated by both 7S and 19S immunoglobulin M (IgM). Reactions proceeded with as little as 0.01 microg of purified 19S or 7S IgM and were complete within 4-6 h. In contrast, purified immunoglobulin did not adsorb to macrophages and had no effect on target cell binding or cytotoxicity. Pretreatment of cells with cytochalasin D abolished the phagocytic reaction, but not spontaneous cytotoxicity. These data show that antibody-mediated killing results from opsonization and phagocytosis; the mechanism of macrophage killing is currently unknown. In addition, these data show that the shark neutrophil, not the macrophage lineage, carries a receptor for Fc mu.


Subject(s)
Antibody-Dependent Cell Cytotoxicity , Immunoglobulin M/physiology , Opsonin Proteins/immunology , Sharks/immunology , Animals , Antibodies, Blocking/pharmacology , Antibodies, Monoclonal/pharmacology , Antibody Specificity , Immunoglobulin Heavy Chains/pharmacology , Immunoglobulin M/biosynthesis , Immunoglobulin M/chemistry , Killer Cells, Natural/immunology , Receptors, Antigen, B-Cell/physiology
17.
Mol Immunol ; 68(2 Pt B): 412-20, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26468036

ABSTRACT

Hottentotta saulcyi, medically important scorpion species, causes some of harmful toxic exposure in Iran. Administrated, conventional antivenom-based immunotherapy is still limited and hardly meet ideal characteristic of effective treatment for scorpion envenomation. In this study we aimed to develop a neutralizing agent directed against scorpion venom based on VHH, variable domain of the Camelidae heavy chain antibody or Nanobody. This promising biomolecule is well-established as an advantageous tool for therapeutic purposes due to its small size, stability, monomeric performance and less immunogenicity. In this study, a large Nb library was constructed and phage displayed after successful camel immunization using H. saulcyi scorpion crude venom. After a series of biopanning rounds on Sephadex G50 purified venom fraction and screening by monoclonal phage ELISA, the best reactive Nb was retrieved and designated Nb12. The selected Nb was then expressed as soluble protein in Escherichia coli, purified and confirmed by SDS-PAGE analysis and western blotting. The lead candidate Nb12 bound scorpion venom with Kaff value of 5×10(7)M(-1). Nb12 was shown to be capable of neutralizing 2 LD50 of whole venom of scorpion toxin when injected in the ratio of the Nb/toxin of 1.4:1 into C57BL/6 mice. In challenge experiment, Nb succeeded to rescue all i.p. lethal dose injected mice even when administrated i.v., 20min after envenoming. These results with ease of production and superior neutralizing activity make Nb a suitable anti-toxin candidate for treatment of scorpion envenoming.


Subject(s)
Antibodies, Neutralizing/immunology , Antivenins/immunology , Camelus/immunology , Scorpion Stings/drug therapy , Scorpion Venoms/antagonists & inhibitors , Single-Domain Antibodies/pharmacology , Animals , Antibody Affinity , Antivenins/pharmacology , Immunization , Immunoglobulin Heavy Chains/immunology , Immunoglobulin Heavy Chains/pharmacology , Immunotherapy/methods , Iran , Mice , Mice, Inbred C57BL , Protein Binding/immunology , Scorpion Stings/pathology , Scorpion Venoms/immunology , Scorpions/metabolism , Single-Domain Antibodies/immunology
18.
J Clin Endocrinol Metab ; 85(3): 1316-9, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10720082

ABSTRACT

Inhibition of tumor necrosis factor (TNF)-alpha results in a marked increase in insulin sensitivity in obese rodents. We investigated the influence of a TNF antagonist [Ro 45-2081, a recombinant fusion protein that consists of the soluble TNF-receptor (p55) linked to the Fc portion of human IgG1] on insulin sensitivity of patients with android obesity. Seven patients (five women and two men; mean +/- SD age, 41 +/- 4 yr; body mass index, 36.1 +/- 4.7 kg/m2; waist to hip ratio, 0.99 +/- 0.11) were studied (three patients with normal glucose tolerance and four patients with impaired glucose tolerance or mild diabetes; all were hyperinsulinemic). Each patient underwent two consecutive euglycemic hyperinsulinemic glucose-clamp tests: 48 h after injection of placebo and 48 h after a single i.v. injection of 50 mg Ro 45-2081. In both tests, steady-state plasma glucose and insulin levels were similar. Insulin-mediated glucose disposal (2.23 +/- 0.74 vs. 2.38 +/- 0.99 mg/kg(-1) x min(-1)) and glucose metabolic clearance rate (2.28 +/- 0.85 vs. 2.48 +/- 1.03 mL/kg(-1) x min(-1)) were similar after placebo and after the drug. Indirect calorimetry showed no difference in substrate oxidation rates between the two experimental conditions. In conclusion, under the conditions of this study, no improvement in insulin sensitivity was observed in obese insulin-resistant patients following a single i.v. administration of a recombinant TNF receptor: Fc fusion protein.


Subject(s)
Immunoglobulin Heavy Chains/analysis , Immunoglobulin Heavy Chains/pharmacology , Insulin Resistance/physiology , Insulin/physiology , Obesity/physiopathology , Receptors, Tumor Necrosis Factor/analysis , Receptors, Tumor Necrosis Factor/physiology , Recombinant Proteins/pharmacology , Adult , Blood Glucose/metabolism , Female , Humans , Hyperinsulinism/blood , Immunoglobulin gamma-Chains , Injections, Intravenous , Insulin/blood , Male , Middle Aged , Recombinant Fusion Proteins/analysis , Recombinant Fusion Proteins/pharmacology , Single-Blind Method , Tumor Necrosis Factor-alpha/pharmacology
19.
Transplantation ; 57(12): 1701-6, 1994 Jun 27.
Article in English | MEDLINE | ID: mdl-8016872

ABSTRACT

The rejection of the transplanted allograft is dependent on T cell activation, which requires T cell receptor engagement by antigen and costimulatory signals delivered by T cell surface molecules such as CD28. CTLA4-Ig is a fusion protein that has previously been shown to block the CD28-mediated costimulatory signal and inhibit immune responses in vitro and in vivo. In this report we show that treatment of the C3H/He recipient of a BALB/c vascularized cardiac allograft with a 12-day course of CTLA4-Ig produced indefinite graft survival (> 100 days) in the majority of recipients. In addition, these recipients demonstrated donor-specific transplantation tolerance when tested with donor-specific (BALB/c) and third-party (C57BL/10) skin grafts. These results demonstrate that CTLA4-Ig can induce transplantation tolerance in the adult murine cardiac allograft model.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antigens, Differentiation/pharmacology , Graft Survival/drug effects , Heart Transplantation/immunology , Immunoconjugates , Abatacept , Animals , Antigens, CD , CTLA-4 Antigen , Humans , Immunoglobulin Heavy Chains/pharmacology , Immunosuppression Therapy/methods , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C3H , Mice, Inbred C57BL , Recombinant Fusion Proteins/pharmacology , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , Time Factors , Transplantation, Homologous/immunology
20.
Peptides ; 21(3): 353-7, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10793216

ABSTRACT

Influence of the ACTH-like peptide H-Val-Lys-Lys-Pro-Gly-Ser-Ser-Val-Lys-Val-OH corresponding to the sequence 11-20 of the variable part of human immunoglobulin G1 (IgG1) heavy chain on growth of MT-4 human T-lymphoblastoid cell line was investigated. It was found that the ACTH-like peptide at concentration range 10(-11) -10(-7) M inhibits the proliferation of MT-4 cells. Labeled ACTH 'address segment' [(125)I]ACTH (13-24) was used to establish that MT-4 cells express specific receptors for ACTH (K(d) = 97 pM). The ACTH-like peptide and human ACTH (but not IgG1 heavy chain) were shown to compete with [(125)I]ACTH (13-24) for binding to these receptors (K(i1) = 0.38 nM and K(i2) = 0.34 nM).


Subject(s)
Adrenocorticotropic Hormone/chemistry , Cell Division/drug effects , Immunoglobulin G/chemistry , Immunoglobulin G/pharmacology , Peptide Fragments/pharmacology , Adrenocorticotropic Hormone/pharmacokinetics , Amino Acid Sequence , Binding, Competitive , Cell Line , Humans , Immunoglobulin Heavy Chains/chemistry , Immunoglobulin Heavy Chains/pharmacology , Kinetics , Peptide Fragments/chemistry , Peptide Fragments/pharmacokinetics , Receptors, Corticotropin/physiology , T-Lymphocytes/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL