Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Gastroenterology ; 153(5): 1351-1362.e4, 2017 11.
Article in English | MEDLINE | ID: mdl-28756234

ABSTRACT

BACKGROUND & AIMS: Although tumor necrosis factor (TNF) antagonists reduce many clinical features of inflammatory bowel disease, complete mucosal healing occurs in fewer than 50% of patients. The Fc-region of monoclonal antibodies against TNF has immunosuppressive properties via effects on macrophage polarization. We examined the interaction between the anti-TNF Fc-region and Fcγ receptors (FcγR), and whether the absence of the Fc core fucose (which increases binding to FcγRIIIa) increases the efficacy of anti-TNF in mice with colitis. METHODS: We generated Rag1-/- mice that lack all activating FcγRs (FcγRI, FcγRIII, and FcγRIV; called FcγR-/-Rag1-/- mice). We produced hypo-fucosylated antibodies against mouse and human TNF (adalimumab). Colitis was induced in mice by transfer of CD4+CD45RBhi to FcγR-/-Rag1-/- or Rag1-/- littermates; mice were given different antibodies against TNF or isotype (control) antibodies and disease activity index scores were determined. Colon tissues were collected and analyzed by histology. Human peripheral blood mononuclear cells (PBMCs) were isolated from blood of healthy donors. T-cell proliferation and proportions of CD206+ (immune regulatory) macrophages were measured in mixed lymphocyte reactions. Human PBMCs were genotyped for FCGR3A158 (the FcγRIIIa-158F allotype displays a lower Fc binding affinity) using the TaqMan single nucleotide polymorphism genotype assay. RESULTS: Rag1-/- mice with colitis given anti-TNF had near complete mucosal healing and Rag1-/- mice given an isotype control antibody developed severe colitis. In contrast, FcγR-/-Rag1-/- mice were refractory to the effects of anti-TNF: their histological colitis scores were as severe as those from FcγR-/-Rag1-/- mice given a control antibody. Colons from Rag1-/- mice that received anti-TNF had an increased number of CD206+ macrophages compared with Rag1-/- mice given control antibody; in FcγR-/-Rag1-/- mice given anti-TNF these numbers were as low as FcγR-/-Rag1-/- given the control antibody. In human PBMCs, anti-TNF increased the number of CD206+ macrophages: this required expression of FcγRIIIa; numbers of these cells were reduced in PBMCs with the low-affinity FcγRIIIa-158F genotype. A hypo-fucosylated form of adalimumab bound human FcγRIIIa with a higher affinity than control adalimumab. When hypo-fucosylated adalimumab was added to PBMCs, a larger number of CD206+ macrophages formed and T-cell proliferation was reduced, compared with addition of a control adalimumab. Hypo-fucosylated adalimumab increased the number of CD206+ macrophages in PMBCs that expressed the low-affinity FcγRIIIa. In mice with colitis, hypo-fucosylated anti-TNF significantly increased the number of CD206+ macrophages in the colon compared with control anti-TNF and was more effective in reducing colitis severity as measured by histology. CONCLUSIONS: In a study of the in vitro and in vivo mechanisms of anti-TNF, we found FcγR engagement by anti-TNF to be required for reduction of colitis in mice and development of CD206+ macrophages. A hypo-fucosylated form of anti-TNF binds FcγRIIIa with higher affinity and induces development of CD206+ macrophages in human PBMCs, especially PBMCs that express low-affinity FcγRIIIa. Hypo-fucosylated anti-TNF might be more effective in patients with inflammatory bowel disease.


Subject(s)
Adalimumab/pharmacology , Antibodies, Monoclonal/pharmacology , Colitis/drug therapy , Colon/drug effects , Immunosuppressive Agents/pharmacology , Intestinal Mucosa/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adoptive Transfer , Animals , Cell Proliferation/drug effects , Cells, Cultured , Colitis/genetics , Colitis/immunology , Colitis/metabolism , Colon/immunology , Colon/metabolism , Colon/pathology , Disease Models, Animal , Genetic Predisposition to Disease , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Lymphocyte Activation/drug effects , Macrophages/drug effects , Macrophages/immunology , Macrophages/metabolism , Mannose Receptor , Mannose-Binding Lectins/immunology , Mannose-Binding Lectins/metabolism , Mice, Inbred C57BL , Mice, Knockout , Phenotype , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Receptors, IgG/deficiency , Receptors, IgG/genetics , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes/transplantation , Time Factors , Tumor Necrosis Factor-alpha/immunology , Wound Healing/drug effects
2.
J Crohns Colitis ; 10(1): 69-76, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26429698

ABSTRACT

BACKGROUND AND AIMS: Anti-tumour necrosis factor [TNF] monoclonal antibodies [infliximab, adalimumab] induce complete mucosal healing in a proportion of patients with Crohn's disease whereas a TNF receptor fusion protein [etanercept] is not effective and the anti-TNF F[ab']2 fragment [certolizumab] shows a very low rate of complete mucosal healing. In contrast, all four TNF-neutralising drugs have demonstrated efficacy in the treatment of rheumatoid arthritis. These observations suggest that factors other than neutralisation of TNF may contribute to clinical outcomes in Crohn's disease. Here we tested the hypothesis that Fc receptor [FcR]-mediated effects may contribute to the therapeutic response of anti-TNF antibodies in inflammatory bowel disease. METHODS: We modified an IgG2c mouse anti-TNF antibody that binds the high-affinity FcRs to generate an IgG1 isotype with strongly diminished binding. We examined the therapeutic effects of both antibodies in the T cell transfer model of inflammatory bowel disease and the collagen-induced arthritis model. RESULTS: The IgG2c anti-TNF antibody prevented colonic inflammation in the T cell transfer model of colitis, whereas the IgG1 anti-TNF did not. Conversely, both the IgG2c and IgG1 anti-TNFs were similarly effective in reducing the severity of articular inflammation in mouse collagen-induced arthritis. CONCLUSION: These data support the concept that the mechanism of action for TNF-neutralising drugs may differ across immune-mediated diseases and, potentially, between therapeutics within a particular disease. Our data suggest a specific role of Fc-mediated immune regulation in the resolution of intestinal inflammation by anti-TNF monoclonal antibodies.


Subject(s)
Antibodies, Monoclonal, Humanized/administration & dosage , Inflammatory Bowel Diseases/drug therapy , Inflammatory Bowel Diseases/immunology , Receptors, Fc/drug effects , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Animals , Arthritis, Experimental/drug therapy , Arthritis, Experimental/immunology , Arthritis, Experimental/pathology , Biomarkers/metabolism , Colitis, Ulcerative/drug therapy , Colitis, Ulcerative/immunology , Colitis, Ulcerative/pathology , Crohn Disease/drug therapy , Crohn Disease/immunology , Crohn Disease/pathology , Disease Models, Animal , Female , Immunohistochemistry , Inflammatory Bowel Diseases/pathology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred DBA , Mice, SCID , Molecular Targeted Therapy/methods , Random Allocation , Receptors, Fc/metabolism , Sensitivity and Specificity , Tumor Necrosis Factor-alpha/administration & dosage
3.
Biotechnol Prog ; 25(3): 735-44, 2009.
Article in English | MEDLINE | ID: mdl-19363814

ABSTRACT

We describe a novel polyprotein precursor-based approach to express antibodies from mammalian cells. Rather than expressing heavy and light chain proteins from separate expression units, the antibody heavy and light chains are contained in one single-open reading frame (sORF) separated by an intein gene fused in frame. Inside mammalian cells this ORF is transcribed into a single mRNA, and translated into one polypeptide. The antibody heavy and light chains are separated posttranslationally, assembled into the functional antibody molecule, and secreted into culture medium. It is demonstrated that Pol I intein from P. horikoshii mediates protein splicing and cleavage reactions in mammalian cells, in the context of antibody heavy and light chain amino acid sequences. To allow the separation of antibody heavy chain, light chain, and the intein, we investigated a number of intein mutations designed to inhibit intein-mediated splicing but preserve cleavage reactions. We have also designed constructs in which the signal peptide downstream from intein has altered hydrophobicity. The use of some of these mutant constructs resulted in more efficient antibody secretion, highlighting areas that can be further explored in improving such an expression system. An antibody secreted using one of the sORF constructs was characterized. This antibody has correct N-terminal sequences for both of its heavy and light chains, correct heavy and light chain MW as well as intact MW as measured by mass spectrometry. Its affinity to antigen, as measured by surface plasmon resonance (SPR), is indistinguishable from that of the same antibody produced using conventional method.


Subject(s)
Antibodies/metabolism , Gene Expression , Open Reading Frames , Polyproteins/metabolism , Protein Engineering/methods , Protein Processing, Post-Translational , Amino Acid Sequence , Antibodies/chemistry , Antibodies/genetics , Archaeal Proteins/genetics , Archaeal Proteins/metabolism , Cell Line , DNA Polymerase I/genetics , DNA Polymerase I/metabolism , Genetic Vectors/genetics , Genetic Vectors/metabolism , Humans , Inteins , Molecular Sequence Data , Polyproteins/chemistry , Polyproteins/genetics , Pyrococcus horikoshii/enzymology , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism
4.
Clin Immunol ; 131(2): 308-16, 2009 May.
Article in English | MEDLINE | ID: mdl-19188093

ABSTRACT

The TNF antagonists adalimumab, infliximab, and etanercept are effective treatments for rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, and psoriasis, but only adalimumab and infliximab have been found to be efficacious in Crohn's disease. The present studies evaluated the TNF-binding and complement-activating properties of adalimumab, infliximab, and etanercept to determine whether these properties may explain differences in their clinical efficacy profiles. Association and dissociation rates of binding to soluble TNF were measured by surface plasmon resonance, and were found to be similar for adalimumab, infliximab, and etanercept, as were their calculated binding affinities. Avidity of binding to soluble TNF, measured by KinExA technology, was 10- to 20-fold greater for soluble etanercept (K(D)=0.4 picomolars [pM]) than for soluble adalimumab or infliximab (K(D)=8.6 and 4.2 pM, respectively). (125)I-adalimumab, -infliximab, and -etanercept bound to membrane TNF (mTNF) on mTNF-transfected cells with similar affinities (K(D)=483, 468, and 445 pM, respectively) that were each lower than for soluble TNF. Complement-dependent cytotoxicity (CDC) was induced in mTNF-transfected cells by adalimumab and infliximab, but was not induced in activated normal human PBMC by any of the 3 agents. In conclusion, the binding properties of adalimumab, infliximab, and etanercept were similar for soluble TNF, and very similar for mTNF, yet none of the 3 was able to induce CDC in activated PBMC. These results suggest that the different clinical efficacy profiles of these agents are not explained by differences in either TNF-intrinsic binding properties or complement lysis.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Antibody Affinity/drug effects , Cell Membrane/immunology , Complement Activation/drug effects , Tumor Necrosis Factor-alpha/metabolism , Adalimumab , Anti-Inflammatory Agents/metabolism , Antibodies, Monoclonal/metabolism , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Etanercept , Humans , Immunoglobulin G/metabolism , Immunoglobulin G/pharmacology , Infliximab , Kinetics , Membrane Proteins/metabolism , Protein Binding/drug effects , Receptors, Tumor Necrosis Factor/metabolism , Solubility , Tumor Necrosis Factor-alpha/antagonists & inhibitors
5.
Pharmacol Ther ; 117(2): 244-79, 2008 Feb.
Article in English | MEDLINE | ID: mdl-18155297

ABSTRACT

During the past 30 years, elucidation of the pathogenesis of rheumatoid arthritis, Crohn's disease, psoriasis, psoriatic arthritis and ankylosing spondylitis at the cellular and molecular levels has revealed that these diseases share common mechanisms and are more closely related than was previously recognized. Research on the complex biology of tumor necrosis factor (TNF) has uncovered many mechanisms and pathways by which TNF may be involved in the pathogenesis of these diseases. There are 3 TNF antagonists currently available: adalimumab, a fully human monoclonal antibody; etanercept, a soluble receptor construct; and infliximab, a chimeric monoclonal antibody. Two other TNF antagonists, certolizumab and golimumab, are in clinical development. The remarkable efficacy of TNF antagonists in these diseases places TNF in the center of our understanding of the pathogenesis of many immune-mediated inflammatory diseases. The purpose of this review is to discuss the biology of TNF and related family members in the context of the potential mechanisms of action of TNF antagonists in a variety of immune-mediated inflammatory diseases. Possible mechanistic differences between TNF antagonists are addressed with regard to their efficacy and safety profiles.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Immune System/drug effects , Inflammation/drug therapy , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Adalimumab , Animals , Anti-Inflammatory Agents/adverse effects , Anti-Inflammatory Agents/chemistry , Anti-Inflammatory Agents/pharmacokinetics , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal, Humanized , Apoptosis/drug effects , Bone and Bones/drug effects , Bone and Bones/immunology , Cartilage/drug effects , Cartilage/immunology , Certolizumab Pegol , Etanercept , Humans , Immune System/metabolism , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin G/pharmacology , Inflammation/immunology , Inflammation/pathology , Infliximab , Ligands , Lymphotoxin-alpha/metabolism , Molecular Structure , Polyethylene Glycols/pharmacology , Receptors, Fc/drug effects , Receptors, Tumor Necrosis Factor , Signal Transduction/drug effects , Structure-Activity Relationship , Tumor Necrosis Factor-alpha/metabolism
7.
Expert Opin Drug Discov ; 2(12): 1549-53, 2007 Dec.
Article in English | MEDLINE | ID: mdl-23488899

ABSTRACT

Biotechnological advances have changed the traditional landscape of the drug discovery process. Mutually beneficial relationships between small, innovative and nimble biotechnology companies and experienced, well-funded pharmaceutical companies have resulted in a more rapid application of new technologies to drug development and diversification. Protein-based drugs, such as therapeutic monoclonal antibodies, exemplify the fulfillment of the promise of the biotechnology revolution.

8.
Curr Protoc Pharmacol ; Chapter 9: Unit 9.7, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-22294129

ABSTRACT

The number of therapeutic antibodies approved by regulatory agencies as novel drugs and the number of antibodies in development has increased significantly. The modular nature of antibody structure has enabled researchers to more predictably design therapeutic antibodies by choosing appropriate functional features most appropriate for a given antibody target and clinical indication. Advances in recombinant antibody technologies have allowed the routine generation of antibodies that can satisfy stringent drug design criteria, such as low immunogenicity, high affinity, target specificity, and commercially viable manufacturing methods. Engineering design opportunities exist for both the variable and the constant regions that encompass, in addition to antigen specificity and affinity, effector functions that mediate immune complex clearance or pharmacokinetics. These are discussed in the context of relevant in vivo and in vitro technologies, such as human IgG transgenic mice, phage display, and biologics manufacturing. Finally, therapeutic antibodies are compared with traditional drugs with respect to target class, selectivity, route of administration, intellectual property issues, and lead discovery and optimization.


Subject(s)
Antibodies/therapeutic use , Drug Discovery/methods , Animals , Antibodies/chemistry , Antibodies/metabolism , Drug Design , Drug Discovery/trends , Half-Life , Humans , Immunogenetic Phenomena/physiology , Immunoglobulins/classification , Mice , Mice, Transgenic , Patents as Topic , Peptide Library , Technology, Pharmaceutical/methods , Technology, Pharmaceutical/trends
9.
N Engl J Med ; 351(20): 2069-79, 2004 Nov 11.
Article in English | MEDLINE | ID: mdl-15537905

ABSTRACT

BACKGROUND: Crohn's disease is associated with excess cytokine activity mediated by type 1 helper T (Th1) cells. Interleukin-12 is a key cytokine that initiates Th1-mediated inflammatory responses. METHODS: This double-blind trial evaluated the safety and efficacy of a human monoclonal antibody against interleukin-12 (anti-interleukin-12) in 79 patients with active Crohn's disease. Patients were randomly assigned to receive seven weekly subcutaneous injections of 1 mg or 3 mg of anti-interleukin-12 per kilogram of body weight or placebo, with either a four-week interval between the first and second injection (Cohort 1) or no interruption between the two injections (Cohort 2). Safety was the primary end point, and the rates of clinical response (defined by a reduction in the score for the Crohn's Disease Activity Index [CDAI] of at least 100 points) and remission (defined by a CDAI score of 150 or less) were secondary end points. RESULTS: Seven weeks of uninterrupted treatment with 3 mg of anti-interleukin-12 per kilogram resulted in higher response rates than did placebo administration (75 percent vs. 25 percent, P=0.03). At 18 weeks of follow-up, the difference in response rates was no longer significant (69 percent vs. 25 percent, P=0.08). Differences in remission rates between the group given 3 mg of anti-interleukin-12 per kilogram and the placebo group in Cohort 2 were not significant at either the end of treatment or the end of follow-up (38 percent and 0 percent, respectively, at both times; P=0.07). There were no significant differences in response rates among the groups in Cohort 1. The rates of adverse events among patients receiving anti-interleukin-12 were similar to those among patients given placebo, except for a higher rate of local reactions at injection sites in the former group. Decreases in the secretion of interleukin-12, interferon-gamma, and tumor necrosis factor alpha by mononuclear cells of the colonic lamina propria accompanied clinical improvement in patients receiving anti-interleukin-12. CONCLUSIONS: Treatment with a monoclonal antibody against interleukin-12 may induce clinical responses and remissions in patients with active Crohn's disease. This treatment is associated with decreases in Th1-mediated inflammatory cytokines at the site of disease.


Subject(s)
Antibodies, Monoclonal/therapeutic use , Crohn Disease/drug therapy , Interleukin-12/immunology , Adult , Aged , Aged, 80 and over , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Colon/immunology , Crohn Disease/immunology , Cytokines/metabolism , Double-Blind Method , Female , Humans , Leukocytes, Mononuclear/metabolism , Male , Middle Aged , Remission Induction
10.
Best Pract Res Clin Rheumatol ; 18(1): 81-95, 2004 Feb.
Article in English | MEDLINE | ID: mdl-15123039

ABSTRACT

Success in drug discovery depends largely on the implementation of appropriate strategies that build on new technologies and the appropriate mix of drug-discovery platforms and research management procedures. Close collaboration between pharmaceutical companies, biotechnology companies and academic institutions during the many intricate phases of drug discovery is necessary to address the need to co-ordinate and streamline target discovery and validation activities, which typically take much longer than anticipated. Antibodies have become an important segment of newly developed therapeutics for a wide range of indications and offer the appropriate risk/benefit profile to balance drug-discovery and development portfolios for optimum success. However, as with other discovery activities, long-term commitment and experience are required to exploit these new techniques fully. Companies with experience in managing the appropriate mix of small-molecule and antibody discovery efforts while implementing novel techniques will remain at the forefront of drug development.


Subject(s)
Antibodies, Monoclonal/pharmacology , Antirheumatic Agents/pharmacology , Biotechnology/standards , Rheumatic Diseases/drug therapy , Adalimumab , Antibodies, Monoclonal, Humanized , Antirheumatic Agents/administration & dosage , Biotechnology/trends , Drug Design , Forecasting , Genomics , Humans , Proteomics , Sensitivity and Specificity , Technology, Pharmaceutical
SELECTION OF CITATIONS
SEARCH DETAIL
...