Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 22
Filter
Add more filters










Publication year range
1.
BMJ Open ; 12(12): e061476, 2022 12 22.
Article in English | MEDLINE | ID: mdl-36549729

ABSTRACT

OBJECTIVES: To assess the risk of new-onset or worsening hyperglycaemia, hypertension, weight gain and hyperlipidaemia with systemic corticosteroid therapy (CST) as reported in published randomised control trial (RCT) studies. DATA SOURCES: Literature search using MEDLINE, EMBASE, Cochrane library, Web of Science and Scopus STUDY ELIGIBILITY CRITERIA: Published articles on results of RCT with a systemic CST arm with numerical data presented on adverse effect (AE). PARTICIPANTS AND INTERVENTIONS: Reports of hyperglycaemia, hypertension, weight gain and hyperlipidaemia associated with systemic CST in patients or healthy volunteer's ≥17 years of age. STUDY APPRAISAL METHODS: Risk of bias tool, assessment at the level of AE and key study characteristics. RESULTS: A total of 5446 articles were screened to include 118 studies with 152 systemic CST arms (total participants=17 113 among which 8569 participants treated with CST). Pooled prevalence of hyperglycaemia in the CST arms within the studies was 10% (95% CI 7% to 14%), with the highest prevalence in respiratory illnesses at 22% (95% CI 9% to 35%). Pooled prevalence of severe hyperglycaemia, hypertension, weight gain and hyperlipidaemia within the corticosteroid arms was 5% (95% CI 2% to 9%), 6% (95% CI 4% to 8%), 13% (95% CI 8% to 18%), 8% (95% CI 4% to 17%), respectively. CST was significantly associated hyperglycaemia, hypertension and weight gain as noted in double-blinded placebo-controlled parallel-arms studies: OR of 2.13 (95% CI 1.66 to 2.72), 1.68 (95% CI 0.96 to 2.95) and 5.20 (95% CI 2.10 to 12.90), respectively. Intravenous therapy posed higher risk than oral therapy: OR of 2.39 (95% CI 1.16 to 4.91). LIMITATIONS: There was significant heterogeneity in the AE definitions and quality of AE reporting in the primary studies and patient populations in the studies. The impact of cumulative dose effect on incidental AE could not be calculated. CONCLUSIONS AND IMPLICATIONS OF KEY FINDINGS: Systemic CST use is associated with increased risk of metabolic AEs, which differs for each disease group and route of administration. PROSPERO REGISTRATION NUMBER: CRD42020161270.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Hyperglycemia , Humans , Adrenal Cortex Hormones/adverse effects , Bias , Hyperglycemia/chemically induced , Hyperglycemia/epidemiology , Hyperglycemia/drug therapy
2.
Am J Respir Crit Care Med ; 206(12): 1463-1479, 2022 12 15.
Article in English | MEDLINE | ID: mdl-35998281

ABSTRACT

Rationale: Idiopathic pulmonary fibrosis (IPF) is a chronic, progressive, and often fatal disorder. Two U.S. Food and Drug Administration-approved antifibrotic drugs, nintedanib and pirfenidone, slow the rate of decline in lung function, but responses are variable and side effects are common. Objectives: Using an in silico data-driven approach, we identified a robust connection between the transcriptomic perturbations in IPF disease and those induced by saracatinib, a selective Src kinase inhibitor originally developed for oncological indications. Based on these observations, we hypothesized that saracatinib would be effective at attenuating pulmonary fibrosis. Methods: We investigated the antifibrotic efficacy of saracatinib relative to nintedanib and pirfenidone in three preclinical models: 1) in vitro in normal human lung fibroblasts; 2) in vivo in bleomycin and recombinant Ad-TGF-ß (adenovirus transforming growth factor-ß) murine models of pulmonary fibrosis; and 3) ex vivo in mice and human precision-cut lung slices from these two murine models as well as patients with IPF and healthy donors. Measurements and Main Results: In each model, the effectiveness of saracatinib in blocking fibrogenic responses was equal or superior to nintedanib and pirfenidone. Transcriptomic analyses of TGF-ß-stimulated normal human lung fibroblasts identified specific gene sets associated with fibrosis, including epithelial-mesenchymal transition, TGF-ß, and WNT signaling that was uniquely altered by saracatinib. Transcriptomic analysis of whole-lung extracts from the two animal models of pulmonary fibrosis revealed that saracatinib reverted many fibrogenic pathways, including epithelial-mesenchymal transition, immune responses, and extracellular matrix organization. Amelioration of fibrosis and inflammatory cascades in human precision-cut lung slices confirmed the potential therapeutic efficacy of saracatinib in human lung fibrosis. Conclusions: These studies identify novel Src-dependent fibrogenic pathways and support the study of the therapeutic effectiveness of saracatinib in IPF treatment.


Subject(s)
Idiopathic Pulmonary Fibrosis , Protein Kinase Inhibitors , Animals , Humans , Mice , Bleomycin/adverse effects , Fibroblasts/metabolism , Fibrosis , Idiopathic Pulmonary Fibrosis/drug therapy , Lung/pathology , Protein Kinase Inhibitors/therapeutic use , src-Family Kinases/metabolism , Transforming Growth Factor beta/metabolism
3.
Clin Exp Allergy ; 52(7): 859-867, 2022 07.
Article in English | MEDLINE | ID: mdl-35524339

ABSTRACT

BACKGROUND: Chronic rhinosinusitis with nasal polyps (CRSwNP) is a type 2 inflammatory disease of the upper airways. AZD1981 is a selective antagonist of chemoattractant receptor-homologous molecule expressed on T helper type 2 and other type 2 cells, including innate lymphoid cells type 2, eosinophils, and basophils. OBJECTIVE: To evaluate the efficacy of AZD1981 in reducing nasal polyp size when added to intranasal corticosteroids in adult patients with CRSwNP. METHODS: Eighty-one subjects (18-70 years of age) with CRSwNP were recruited and screened for trial eligibility from allergy and otolaryngology clinics from a single tertiary care site between June 2016 and August 2019. Eligible patients were randomized in a double-blind fashion to receive either AZD1981 (n = 22) or placebo (n = 21) orally three times a day for 12 weeks, added to intranasal corticosteroids. The primary endpoint was a change in nasal polyp score (NPS) at 12 weeks. Secondary endpoints included improvement in sinus computed tomography using Lund Mackay scoring, symptoms using visual analog scale, quality of life using Sino Nasal Outcome Test-22, and the Brief Smell Identification Test. RESULTS: Forty-three patients met the inclusion criteria and were enrolled. At 12 weeks, there was no difference in NPS change in the AZD1981 arm (mean 0, standard error 0.34, n = 15) compared with placebo (mean 0.20, standard error 0.36, n = 17); mean difference -0.20 (95% confidence interval: -1.21, 0.81; p = .69). No significant differences were observed for Lund Mackay score, symptoms, quality of life, or smell test. AZD1981 was well tolerated except for one case of hypersensitivity reaction. CONCLUSION: In patients with CRSwNP, the addition of AZD1981 to intranasal corticosteroids did not change nasal polyp size, radiographic scores, symptoms, or disease-specific quality of life.


Subject(s)
Nasal Polyps , Rhinitis , Sinusitis , Acetates , Adrenal Cortex Hormones/therapeutic use , Adult , Chronic Disease , Humans , Immunity, Innate , Indoles , Lymphocytes , Nasal Polyps/complications , Nasal Polyps/drug therapy , Quality of Life , Rhinitis/complications , Rhinitis/drug therapy , Sinusitis/drug therapy
4.
Sci Rep ; 11(1): 20827, 2021 10 21.
Article in English | MEDLINE | ID: mdl-34675338

ABSTRACT

Non-alcoholic steatohepatitis (NASH) is a rising health challenge, with no approved drugs. We used a computational drug repositioning strategy to uncover a novel therapy for NASH, identifying a GABA-B receptor agonist, AZD3355 (Lesogaberan) previously evaluated as a therapy for esophageal reflux. AZD3355's potential efficacy in NASH was tested in human stellate cells, human precision cut liver slices (hPCLS), and in vivo in a well-validated murine model of NASH. In human stellate cells AZD3355 significantly downregulated profibrotic gene and protein expression. Transcriptomic analysis of these responses identified key regulatory nodes impacted by AZD3355, including Myc, as well as MAP and ERK kinases. In PCLS, AZD3355 down-regulated collagen1α1, αSMA and TNF-α mRNAs as well as secreted collagen1α1. In vivo, the drug significantly improved histology, profibrogenic gene expression, and tumor development, which was comparable to activity of obeticholic acid in a robust mouse model of NASH, but awaits further testing to determine its relative efficacy in patients. These data identify a well-tolerated clinical stage asset as a novel candidate therapy for human NASH through its hepatoprotective, anti-inflammatory and antifibrotic mechanisms of action. The approach validates computational methods to identify novel therapies in NASH in uncovering new pathways of disease development that can be rapidly translated into clinical trials.


Subject(s)
Drug Repositioning , GABA-B Receptor Agonists/therapeutic use , Liver/drug effects , Non-alcoholic Fatty Liver Disease/drug therapy , Phosphinic Acids/therapeutic use , Propylamines/therapeutic use , Adult , Aged , Animals , Cell Line , Disease Models, Animal , Female , GABA-B Receptor Agonists/pharmacology , Humans , Liver/metabolism , Liver/pathology , Male , Mice , Mice, Inbred C57BL , Middle Aged , Non-alcoholic Fatty Liver Disease/metabolism , Non-alcoholic Fatty Liver Disease/pathology , Phosphinic Acids/pharmacology , Propylamines/pharmacology
5.
Hum Immunol ; 75(12): 1139-46, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25454619

ABSTRACT

IVIG is frequently used in the 'pre-conditioning' regimens for higher risk transplants; its effects are attributed in part to induction of Tregs. We have identified regulatory T cell (Treg) epitopes, now known as Tregitopes, in IgG, the main component of intravenous immunoglobulin therapy (IVIg). Tregitopes provide one explanation for the expansion and activation of Treg cells following IVIg treatment. Tregitopes are peptides that exhibit high affinity binding to multiple human HLA Class II DR; they are conserved across IgG isotypes and mammalian species. In vitro and in vivo, for human PBMC and in animal models, Tregitopes activate Tregs. Studies to delineate the mechanism of action have shown that Tregitopes' effects are very similar to IVIg in vitro. Here we demonstrate that Tregitopes induce Tregs to produce IL-10, leading to modulation of dendritic cell phenotype (down-regulation of Class II, CD80 and CD86 and up-regulation of ILT3), and describe the effects of Tregitopes in the ABM-TCR-transgenic skin transplantation model. The discovery of Tregitopes in IgG and other autologous proteins may contribute to improved understanding of the mechanism of action of IVIg and lead to the application of these powerful immunomodulators to improve transplantation success and suppress autoimmune disease, in the future.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunoglobulin G/immunology , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology , Transplantation Tolerance/immunology , Animals , B7-1 Antigen/biosynthesis , B7-2 Antigen/biosynthesis , Dendritic Cells/immunology , Female , HLA-DR Antigens/immunology , Humans , Immune Tolerance/immunology , Immunization, Passive , Immunoglobulins, Intravenous , Immunomodulation , Interleukin-10/biosynthesis , Leukocytes, Mononuclear/immunology , Membrane Glycoproteins , Mice , Mice, Inbred C57BL , Receptors, Cell Surface/biosynthesis , Receptors, Immunologic , Skin Transplantation
6.
Clin Immunol ; 149(3): 534-55, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24263283

ABSTRACT

Protein therapeutics hold a prominent and rapidly expanding place among medicinal products. Purified blood products, recombinant cytokines, growth factors, enzyme replacement factors, monoclonal antibodies, fusion proteins, and chimeric fusion proteins are all examples of therapeutic proteins that have been developed in the past few decades and approved for use in the treatment of human disease. Despite early belief that the fully human nature of these proteins would represent a significant advantage, adverse effects associated with immune responses to some biologic therapies have become a topic of some concern. As a result, drug developers are devising strategies to assess immune responses to protein therapeutics during both the preclinical and the clinical phases of development. While there are many factors that contribute to protein immunogenicity, T cell- (thymus-) dependent (Td) responses appear to play a critical role in the development of antibody responses to biologic therapeutics. A range of methodologies to predict and measure Td immune responses to protein drugs has been developed. This review will focus on the Td contribution to immunogenicity, summarizing current approaches for the prediction and measurement of T cell-dependent immune responses to protein biologics, discussing the advantages and limitations of these technologies, and suggesting a practical approach for assessing and mitigating Td immunogenicity.


Subject(s)
Biological Products/immunology , Immunity, Cellular/drug effects , T-Lymphocytes/drug effects , Animals , Antibodies, Monoclonal/administration & dosage , Antibodies, Monoclonal/immunology , Antibody Formation , B-Lymphocytes/drug effects , B-Lymphocytes/immunology , Biological Assay , Biological Products/administration & dosage , Biomarkers, Pharmacological/analysis , Cytokines/administration & dosage , Cytokines/immunology , Drug Evaluation, Preclinical , Humans , Intercellular Signaling Peptides and Proteins/administration & dosage , Intercellular Signaling Peptides and Proteins/immunology , Molecular Docking Simulation , Recombinant Proteins/administration & dosage , Recombinant Proteins/immunology , T-Lymphocytes/immunology
7.
Expert Rev Clin Pharmacol ; 6(6): 651-62, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24164613

ABSTRACT

Immune responses to some monoclonal antibodies (mAbs) and biologic proteins interfere with their efficacy due to the development of anti-drug antibodies (ADA). In the case of mAbs, most ADA target 'foreign' sequences present in the complementarity determining regions (CDRs). Humanization of the mAb sequence is one approach that has been used to render biologics less foreign to the human immune system. However, fully human mAbs can also drive immunogenicity. De-immunization (removing epitopes) has been used to reduce biologic protein immunogenicity. Here, we discuss a third approach to reducing the immunogenicity of biologics: introduction of Treg epitopes that stimulate Treg function and induce tolerance to the biologic protein. Supplementing humanization (replacing xeno-sequences with human) and de-immunization (reducing T effector epitopes) with tolerization (introducing Treg epitopes) where feasible, as a means of improving biologics 'quality by design', may lead to the development of ever more clinically effective, but less immunogenic, biologics.


Subject(s)
Biological Products/adverse effects , Animals , Antibodies, Monoclonal/adverse effects , Antibodies, Monoclonal/immunology , Antibody Affinity , Antibody Specificity , Epitopes, T-Lymphocyte , Humans
8.
Mol Ther ; 21(9): 1727-37, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23857231

ABSTRACT

Immune responses directed against viral capsid proteins constitute a main safety concern in the use of adeno-associated virus (AAV) as gene transfer vectors in humans. Pharmacological immunosuppression has been proposed as a solution to the problem; however, the approach suffers from several potential limitations. Using MHC class II epitopes initially identified within human IgG, named Tregitopes, we showed that it is possible to modulate CD8+ T cell responses to several viral antigens in vitro. We showed that incubation of peripheral blood mononuclear cells with these epitopes triggers proliferation of CD4+CD25+FoxP3+ T cells that suppress killing of target cells loaded with MHC class I antigens in an antigen-specific fashion, through a mechanism that seems to require cell-to-cell contact. Expression of a construct encoding for the AAV capsid structural protein fused to Tregitopes resulted in reduction of CD8+ T cell reactivity against the AAV capsid following immunization with an adenoviral vector expressing capsid. This was accompanied by an increase in frequency of CD4+CD25+FoxP3+ T cells in spleens and lower levels of inflammatory infiltrates in injected tissues. This proof-of-concept study demonstrates modulation of CD8+ T cell reactivity to an antigen using regulatory T cell epitopes is possible.


Subject(s)
Antigens, Viral/immunology , CD8-Positive T-Lymphocytes/immunology , Capsid Proteins/immunology , Dependovirus/immunology , Epitopes, T-Lymphocyte/immunology , Genetic Vectors , Immunoglobulin G/immunology , Animals , Antigens, Viral/genetics , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Capsid/immunology , Capsid Proteins/genetics , Capsid Proteins/metabolism , Cells, Cultured , Dependovirus/genetics , Epitopes, T-Lymphocyte/genetics , Epitopes, T-Lymphocyte/metabolism , Genetic Therapy , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class II/immunology , Humans , Immunoglobulin G/genetics , Male , Mice , Mice, Inbred C57BL , Spleen/immunology , T-Lymphocytes, Regulatory/immunology
9.
R I Med J (2013) ; 96(2): 19-21, 2013 Feb 01.
Article in English | MEDLINE | ID: mdl-23641421

ABSTRACT

EpiVax, Inc., is an early-stage informatics and immunology biotechnology company in Providence, Rhode Island. It applies computational tools to harness immunity in three major areas: immunomodulation, biotherapeutic immunogenicity risk assessment and de-risking, and vaccine development. Immunotherapy, bio-better and vaccine candidates under development at EpiVax promise to improve the health outcomes of millions of people affected by devastating immune-related diseases.


Subject(s)
Autoimmune Diseases/immunology , Biomedical Research/trends , Computational Biology , Drug Discovery/trends , Drug Industry , Immunodominant Epitopes/immunology , Immunomodulation , T-Lymphocytes, Regulatory/immunology , Autoimmune Diseases/drug therapy , Autoimmunity , Drug Design , Humans , Rhode Island , Vaccines
10.
J Diabetes Res ; 2013: 621693, 2013.
Article in English | MEDLINE | ID: mdl-23710469

ABSTRACT

HLA class II-restricted regulatory T cell (Treg) epitopes in IgG (also called "Tregitopes") have been reported to suppress immune responses to coadministered antigens by stimulating the expansion of natural Tregs (nTregs). Here we evaluate their impact on human immune responses to islet cell antigens ex vivo and on the modulation of type 1 diabetes (T1D) in a murine model in vivo. Co-administration of Tregitopes and T1D antigens delayed development of hyperglycemia and reduced the incidence of diabetes in NOD mice. Suppression of diabetes could be observed even following onset of disease. To measure the impact of Tregitope treatment on T cell responses, we evaluated the effect of Tregitope treatment in DO11.10 mice. Upregulation of FoxP3 in KJ1-26-stained OVA-specific CD4(+) T cells was observed following treatment of DO11.10 mice with Tregitopes, along with reductions in anti-OVA Ig and T effector responses. In ex vivo studies of human T cells, peripheral blood mononuclear cells' (PBMC) responses to GAD65 epitopes in the presence and absence of Tregitope were variable. Suppression of immune responses to GAD65 epitopes ex vivo by Tregitope appeared to be more effective in assays using PBMC from a newly diagnosed diabetic subject than for other more established diabetic subjects, and correlation of the degree of suppression with predicted HLA restriction of the Tregitopes was confirmed. Implementation of these defined regulatory T cell epitopes for therapy of T1D and other autoimmune diseases may lead to a paradigm shift in disease management.


Subject(s)
Diabetes Mellitus, Type 1/immunology , Epitopes, T-Lymphocyte/immunology , Immune Tolerance/immunology , Immunoglobulin G/immunology , T-Lymphocytes, Regulatory/immunology , Adaptive Immunity/immunology , Alleles , Animals , Cell Proliferation , Diabetes Mellitus, Type 1/genetics , HLA-DR4 Antigen/genetics , HLA-DR4 Antigen/immunology , Humans , Immunophenotyping , Mice , Mice, Inbred NOD
11.
J Clin Immunol ; 33 Suppl 1: S43-9, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22941509

ABSTRACT

Tregitopes are regulatory T cell epitopes derived from immunoglobulin G (IgG) that stimulate CD25(+) FoxP3(+) T cells to expand. In conjunction with these Tregs, Tregitopes can prevent, treat, and even cure autoimmune disease in mouse models, suppress allo-specific responses in murine transplant models, inhibit CD8(+) T cell responses to recombinant adeno-associated virus (AAV) gene transfer vectors, and induce adaptive Tregs in DO11.10 mice. In this review of recent Tregitope studies, we summarize their effects in vitro and describe recent comparisons between intravenous IgG (IVIG) and Tregitopes in standard in vivo immune tolerance models. Further investigations of the mechanism of action of Tregitopes in the preclinical models described here will lead to clinical trials where Tregitopes may have the potential to alter the treatment of autoimmune disease, transplantation, and allergy, and to improve the efficiency of gene and protein replacement therapies.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunoglobulin G/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/therapy , Autoimmunity , Humans , Immune Tolerance , Immunoglobulins, Intravenous/immunology , Immunoglobulins, Intravenous/therapeutic use , Research/trends
12.
Autoimmun Rev ; 12(3): 436-43, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22944299

ABSTRACT

In the course of screening immunoglobulin G (IgG) sequences for T cell epitopes, we identified novel Treg epitope peptides, now called Tregitopes, contained in the highly conserved framework regions of Fab and Fc. Tregitopes may provide one explanation for the expansion and stimulation of Treg cells following intravenous immunoglobulin (IVIg) therapy. Their distinguishing characteristics include in silico signatures that suggest high-affinity binding to multiple human HLA class II DR and conservation across IgG isotypes and mammalian species with only minor amino acid modifications. Tregitopes induce expansion of CD4(+)/CD25(hi)/FoxP3(+) T cells and suppress immune responses to co-incubated antigens in vitro. By comparing the human IgG Tregitopes (hTregitopes 167 and 289, located in the IgG CH1 and CH2 domains) and Fab to murine sequences, we identified class II-restricted murine Tregitope homologs (mTregitopes). In vivo, mTregitopes suppress inflammation and reproducibly induce Tregs to expand. In vitro studies suggest that the Tregitope mechanism of action is to induce Tregs to respond, leading to production of regulatory signals, followed by modulation of dendritic cell phenotype. The identification of Treg epitopes in IgG suggests that additional Tregitopes may also be present in other autologous proteins; methods for identifying and validating such peptides are described here. The discovery of Tregitopes in IgG and other autologous proteins may lead to the development of new insights as to the role of Tregs in autoimmune diseases.


Subject(s)
Epitopes, T-Lymphocyte/immunology , Immunoglobulins, Intravenous/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Epitope Mapping , Epitopes, T-Lymphocyte/metabolism , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class II/metabolism , Humans , Immunoglobulin G/immunology , Immunoglobulin G/metabolism , Immunoglobulins, Intravenous/metabolism , Mice , Protein Binding
13.
Clin Dev Immunol ; 2013: 493138, 2013.
Article in English | MEDLINE | ID: mdl-24454476

ABSTRACT

Five years ago, we reported the identification and characterization of several regulatory T-cell epitopes (now called Tregitopes) that were discovered in the heavy and light chains of IgG (De Groot et al. Blood, 2008). When added ex vivo to human PBMCs, these Tregitopes activated regulatory T cells (Tregs), increased expression of the transcription factor FoxP3, and induced IL-10 expression in CD4(+) T cells. We have now shown that coadministration of the Tregitopes in vivo, in a number of different murine models of autoimmune disease, can suppress immune responses to antigen in an antigen-specific manner, and that this response is mediated by Tregs. In addition we have shown that, although these are generally promiscuous epitopes, the activity of individual Tregitope peptides is restricted by HLA. In this brief report, we provide an overview of the effects of Tregitopes in vivo, discuss potential applications, and suggest that Tregitopes may represent one of the "active pharmaceutical ingredients" of IVIg. Tregitope applications may include any of the autoimmune diseases that are currently treated almost exclusively with intravenous immunoglobulin G (IVIG), such as Chronic Inflammatory Demyelinating Polyneuropathy (CIDP) and Multifocal Motor Neuropathy (MMN), as well as gene therapy and allergy where Tregitopes may provide a means of inducing antigen-specific tolerance.


Subject(s)
Epitopes, T-Lymphocyte/administration & dosage , Peptides/administration & dosage , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/immunology , Animals , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Epitopes, T-Lymphocyte/immunology , Humans , Immune Tolerance/drug effects , Immunoglobulins, Intravenous/immunology , Immunoglobulins, Intravenous/therapeutic use , Lymphocyte Activation/drug effects , Lymphocyte Activation/immunology , Peptides/immunology , T-Lymphocytes, Regulatory/metabolism
14.
Hum Vaccin Immunother ; 8(10): 1459-64, 2012 Oct.
Article in English | MEDLINE | ID: mdl-23095864

ABSTRACT

Babies born with Pompe disease require life-long treatment with enzyme-replacement therapy (ERT). Despite the human origin of the therapy, recombinant human lysosomal acid α glucosidase (GAA, rhGAA), ERT unfortunately leads to the development of high titers of anti-rhGAA antibody, decreased effectiveness of ERT, and a fatal outcome for a significant number of children who have Pompe disease. The severity of disease, anti-drug antibody (ADA) development, and the consequences thereof are directly related to the degree of the enzyme deficiency. Babies born with a complete deficiency GAA are said to have cross-reactive immunologic material (CRIM)-negative Pompe disease and are highly likely to develop GAA ADA. Less frequently, GAA ADA develop in CRIM-positive individuals. Currently, GAA-ADA sero-positive babies are treated with a combination of immunosuppressive drugs to induce immunological tolerance to ERT, but the long-term effect of these regimens is unknown. Alternative approaches that might redirect the immune response toward antigen-specific tolerance without immunosuppressive agents are needed. Methods leading to the induction of antigen-specific regulatory T cells (Tregs), using peptides such as Tregitopes (T regulatory cell epitopes) are under consideration for the future treatment of CRIM-negative Pompe disease. Tregitopes are natural T cell epitopes derived from immunoglobulin G (IgG) that cause the expansion and activation of regulatory T cells (Treg). Teaching the immune system to tolerate GAA by co-delivering GAA with Tregitope peptides might dramatically improve the lives of CRIM-negative babies and could be applied to other enzyme replacement therapies to which ADA have been induced.


Subject(s)
Enzyme Replacement Therapy/methods , Glycogen Storage Disease Type II/therapy , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/immunology , Humans , Immunosuppressive Agents/therapeutic use , Lysosomes/enzymology , Models, Biological , alpha-Glucosidases/immunology , alpha-Glucosidases/therapeutic use
15.
Biologicals ; 40(5): 364-8, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22770604

ABSTRACT

Protein therapeutics occupy a very significant position in the biopharmaceutical market. In addition to the preclinical, clinical and post marketing challenges common to other drugs, unwanted immunogenicity is known to affect efficacy and/or safety of most biotherapeutics. A standard set of immunogenicity risk factors are routinely used to inform monitoring strategies in clinical studies. A number of in-silico, in vivo and in vitro approaches have also been employed to predict immunogenicity of biotherapeutics, but with limited success. Emerging data also indicates the role of immune tolerance mechanisms and impact of several product-related factors on modulating host immune responses. Thus, a comprehensive discussion of the impact of innate and adaptive mechanisms and molecules involved in induction of host immune responses on immunogenicity of protein therapeutics is needed. A detailed understanding of these issues is essential in order to fully exploit the therapeutic potential of this class of drugs. This Roundtable Session was designed to provide a common platform for discussing basic immunobiological and pharmacological issues related to the role of biotherapeutic-associated risk factors, as well as host immune system in immunogenicity against protein therapeutics. The session included overview presentations from three speakers, followed by a panel discussion with audience participation.


Subject(s)
Adaptive Immunity/drug effects , Proteins/immunology , Proteins/therapeutic use , Adaptive Immunity/physiology , Animals , Antibodies/analysis , Antibodies/immunology , Antibody Formation/drug effects , Biological Products/immunology , Biological Products/therapeutic use , Drug Antagonism , Drug Compounding/adverse effects , Drug Contamination , Humans , Immunoglobulins/chemistry , Immunoglobulins/immunology , Immunoglobulins/therapeutic use , Mice , Pharmaceutical Preparations , Quality Control , Recombinant Proteins/immunology , Recombinant Proteins/therapeutic use
16.
Expert Opin Drug Discov ; 6(1): 9-15, 2011 Jan.
Article in English | MEDLINE | ID: mdl-22646824

ABSTRACT

The role of cellular immunity as a mediator of protection against disease is gaining recognition, particularly with regard to the many pathogens for which we presently lack effective vaccines. As a result, there is an ever-increasing need to understand the T-cell populations induced by vaccination and, therefore, T-cell epitopes responsible for triggering their activation. Although the characterization and harnessing of cellular immunity for vaccine development is an active area of research interest, the field still needs to rigorously define T-cell epitope specificities, above all, on a genomic level. New immunoinformatic epitope mapping tools now make it possible to identify pathogen epitopes and perform comparisons against human and microbial genomic data sets. Such information will help to determine whether adaptive immune responses elicited by a vaccine are both pathogen-specific and protective, but not crossreactive against host or host-associated sequences that could jeopardize self-tolerance and/or human microbiome-host homeostasis. Here, we discuss advances in genomics and vaccine design and their relevance to the development of safer, more effective vaccines.

17.
J Biol Chem ; 281(26): 17707-17, 2006 Jun 30.
Article in English | MEDLINE | ID: mdl-16636048

ABSTRACT

Stat3 and its isoforms belong to a family of cytoplasmic transcription factors that affect the synthesis of various proteins. Caspases are cysteinyl-aspartate proteases that function under apoptotic and non-apoptotic conditions. We now report that, in addition to transcriptional splicing, Stat3 fragmentation can be mediated by caspases. Caspase activation in DU145 cells was achieved by staurosporine (STS) exposure, and Western analysis revealed a reduction in full-length Stat3 (fl-Stat3) expression that was caspase-mediated. This proteolytic relationship was further studied by exposing purified Stat3 protein to a mixture of active caspases under cell-free conditions. This demonstrated that caspases directly cleaved Stat3 and Stat3 cleavage was accompanied by the apparent formation of cleavage fragment(s). Stat3 cleavage fragments, reflecting multiple caspase cleavage sites, also were observed in vitro following STS exposure in DU145 cells and in HEK293T cells transfected to express Stat3 truncation mutants. The impact of cleavage on Stat3 transcriptional activity next was assessed and revealed that cleavage of fl-Stat3 was accompanied by reductions in Stat3-DNA binding, Stat3-driven reporter protein (luciferase) activity, and the expression of selected Stat3-dependent genes. Further, reduced Stat3 expression correlated with increased sensitivity to apoptotic stimuli. In concomitant experiments, reporter activity was assessed in Stat3 truncation mutant-expressing HEK293T cells and revealed that, under non-apoptotic conditions, expression of different Stat3 fragments induced differential effects on Stat3-driven luciferase activity. These findings demonstrate that fl-Stat3 undergoes proteolytic processing by caspases that reduces its expression and leads to the formation of cleavage fragments that may modulate Stat3 transcriptional activity.


Subject(s)
Apoptosis/physiology , Caspases/metabolism , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , Transcriptional Activation/physiology , Amino Acid Chloromethyl Ketones/pharmacology , Apoptosis/drug effects , Caspase Inhibitors , Cysteine Proteinase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Gene Expression Regulation/physiology , HeLa Cells , Humans , Mutagenesis , Peptide Fragments/genetics , Peptide Fragments/metabolism , Staurosporine/pharmacology
18.
Clin Cancer Res ; 12(2): 569-76, 2006 Jan 15.
Article in English | MEDLINE | ID: mdl-16428502

ABSTRACT

PURPOSE: Cancer immunotherapy has been limited by anergy of patient T cells, inadequate numbers of precursor tumor-specific CTL, and difficulty in producing therapeutic doses of CTL. To overcome these limitations, bispecific antibodies have been used to create artificial antibody receptors that direct polyclonal activated T cells (ATC) to target tumor antigens. Studies reported herein were designed to characterize bispecific antibody-armed ATC functions during multiple rounds of targeted cell stimulation. EXPERIMENTAL DESIGN: ATCs were generated from human peripheral blood mononuclear cells (PBMC) by culture with anti-CD3 and interleukin 2 for 14 days and armed with anti-CD3 x anti-Her2 bispecific antibody (Her2Bi). In vitro, Her2Bi-armed ATC were examined for a range of functions after repeated stimulation with the Her2/neu-expressing breast cancer cell line SK-BR-3. PBMC isolated from cancer patients treated with Her2Bi-armed ATC were tested ex vivo for cytotoxicity against SK-BR-3. RESULTS: In vitro, armed ATC divided, maintained surface Her2Bi, and expressed a range of activities for extended periods of time. Perforin-mediated cytotoxic activity by armed ATC continued for at least 336 hours, and cytokines and chemokines (i.e., IFN-gamma and regulated on activation, normal T-cell expressed and secreted protein [RANTES]) were secreted during successive rounds of stimulation. Furthermore, PBMC isolated from patients over their courses of immunotherapy exhibited significant cytolytic activity against SK-BR-3 as a function of Her2Bi-armed ATC infusions. CONCLUSIONS: These studies show that armed ATC are specific, durable, and highly functional T-cell populations in vitro. These previously unappreciated broad and long-term functions of armed ATC are encouraging for their therapeutic use in treating cancer.


Subject(s)
Antibodies, Bispecific/pharmacology , Breast Neoplasms/therapy , CD3 Complex/immunology , Cytotoxicity, Immunologic , Receptor, ErbB-2/immunology , T-Lymphocytes/immunology , Breast Neoplasms/immunology , Cell Line, Tumor , Chemokine CCL5/metabolism , Female , Humans , Immunotherapy , Interferon-gamma/metabolism , Interleukin-2/immunology , Lymphocyte Activation , Membrane Glycoproteins/pharmacology , Perforin , Pore Forming Cytotoxic Proteins
19.
J Immunol ; 174(1): 320-7, 2005 Jan 01.
Article in English | MEDLINE | ID: mdl-15611255

ABSTRACT

There is a growing appreciation for how cells integrate and appropriately respond to competing signals for proliferation and apoptosis. The studies described in this report examined the effects of exposure to the cytokine IFN-alpha (IFN-alpha-2a) on sensitivity of the human cell lines H9 and SKW6.4 to Fas ligand (FasL)-induced apoptosis. In a concentration-dependent manner, FasL induced apoptosis, as shown by Western blot analysis of procaspase 8 and poly(ADP-ribose) polymerase cleavage after 3-h exposure and by cytofluorometric analysis of sub-G(0)-G(1) cellular DNA content after 24-h exposure. H9 and SKW6.4 cell lines responded to 10,000 IU/ml IFN-alpha-2a, as evidenced by decreased cell proliferation and tyrosine phosphorylation of Stat1 and Stat3 proteins without significant cytotoxicity. The effects of cytokine exposure on apoptosis were examined; incubation in medium containing 10,000 IU/ml IFN-alpha-2a for 1 h before FasL treatment significantly reduced all above-mentioned hallmarks of apoptosis. Surprisingly, these antiapoptotic effects of IFN-alpha-2a were independent of de novo protein synthesis, because they occurred in both the absence and the presence of cycloheximide. However, chemical inhibitors of JAK completely abrogated the effects of IFN-alpha-2a on FasL-induced apoptosis, indicating a direct role for JAK-mediated protein phosphorylation in modulating sensitivity to apoptosis. Together, these data suggest a novel mechanism, independent of protein synthesis, by which cytokine signals through JAKs can interact with and attenuate this receptor-mediated apoptotic process.


Subject(s)
Apoptosis/drug effects , Membrane Glycoproteins/pharmacology , Protein-Tyrosine Kinases/metabolism , Signal Transduction/physiology , Apoptosis/immunology , Blotting, Western , Cell Proliferation/drug effects , Cycloheximide/pharmacology , DNA-Binding Proteins/drug effects , DNA-Binding Proteins/metabolism , Dose-Response Relationship, Drug , Fas Ligand Protein , Humans , Interferon-alpha/immunology , Interferon-alpha/pharmacology , Janus Kinase 1 , Membrane Glycoproteins/immunology , Phosphorylation , Protein Synthesis Inhibitors/pharmacology , Protein-Tyrosine Kinases/immunology , STAT1 Transcription Factor , STAT3 Transcription Factor , Trans-Activators/drug effects , Trans-Activators/metabolism , Tumor Cells, Cultured , Tyrosine/drug effects , Tyrosine/metabolism
20.
Cancer Chemother Pharmacol ; 54(3): 249-58, 2004 Sep.
Article in English | MEDLINE | ID: mdl-15173956

ABSTRACT

Taurolidine (TRD) was designed in the 1970s as a broad-spectrum antibiotic and is used clinically at high doses without systemic toxicity. We have found that this agent possesses cytotoxic activity in human tumor cell lines and antineoplastic activity in mice bearing i.p. human tumor xenografts. We now report the mechanism by which TRD induces cell death in DU145 human prostate tumor cells. The IC50 (3 days) of TRD in this model was 16.8+/-1.1 microM. Cytotoxicity was associated with DNA debris and increased membrane phosphatidylserine externalization, both suggesting the induction of apoptosis. This was confirmed by the ability of TRD to induce PARP cleavage in these cells, an effect prevented by coexposure to the pan-caspase inhibitor zVAD-FMK. TRD exposure also resulted in the appearance of cytochrome c in the cytoplasm, procaspase 9 activation within 2 h of drug exposure and procaspase 8 activation 4 h after exposure. Parallel experiments revealed that cytochrome c appearance in the cytoplasm was not blocked by preexposure to zVAD-FMK, while activation of both procaspase 9 and procaspase 8 was prevented. Finally, antineoplastic activity was assessed in mice bearing subcutaneous xenografts of DU145 cells. Initial studies quantitated the toxicity of three i.p. injections of TRD, administered as one injection on three alternate days per week, at doses ranging from 500 to 700 mg/kg per injection. The 500 mg/kg dose produced about 7% mortality after three cycles and effectively inhibited tumor growth. Thus, TRD induced mitochondrial-mediated apoptosis in DU145 human prostate tumor cells and this effect could be exploited for therapeutic advantage.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Prostatic Neoplasms/drug therapy , Taurine/analogs & derivatives , Taurine/pharmacology , Thiadiazines/pharmacology , Animals , Antineoplastic Agents/administration & dosage , Humans , Injections, Intraperitoneal , Male , Mice , Mice, Nude , Mitochondria/drug effects , Mitochondria/pathology , Neoplasms, Experimental , Prostatic Neoplasms/pathology , Prostatic Neoplasms/veterinary , Taurine/administration & dosage , Thiadiazines/administration & dosage , Transplantation, Heterologous , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...