Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 181
Filter
1.
Biomolecules ; 11(12)2021 11 23.
Article in English | MEDLINE | ID: mdl-34944387

ABSTRACT

Breastfeeding-or lactation-is a unique and defining reproductive trait of mammals that nourishes offspring by supplying nutrient-rich breast milk [...].


Subject(s)
Biological Factors/chemistry , Breast Feeding/statistics & numerical data , Milk, Human/chemistry , Biological Factors/immunology , Breast Feeding/trends , Child Development , Female , Homeostasis , Humans , Immune System/growth & development , Infant , Milk, Human/immunology
2.
Biomolecules ; 11(12)2021 12 18.
Article in English | MEDLINE | ID: mdl-34944544

ABSTRACT

The gastrointestinal system is responsible for the digestion and the absorption of nutrients. At the same time, it is essentially involved in the maintenance of immune homeostasis. The strongest antigen contact in an organism takes place in the digestive system showing the importance of a host to develop mechanisms allowing to discriminate between harmful and harmless antigens. An efficient intestinal barrier and the presence of a large and complex part of the immune system in the gut support the host to implement this task. The continuous ingestion of harmless antigens via the diet requires an efficient immune response to reliably identify them as safe. However, in some cases the immune system accidentally identifies harmless antigens as dangerous leading to various diseases such as celiac disease, inflammatory bowel diseases and allergies. It has been shown that the intestinal immune function can be affected by bioactive compounds derived from the diet. The present review provides an overview on the mucosal immune reactions in the gut and how bioactive food ingredients including secondary plant metabolites and probiotics mediate its health promoting effects with regard to the intestinal immune homeostasis.


Subject(s)
Biological Factors/administration & dosage , Immunity , Intestinal Mucosa/immunology , Animals , Biological Factors/immunology , Diet/classification , Humans , Phytochemicals/administration & dosage , Phytochemicals/immunology , Prebiotics/administration & dosage , Probiotics/administration & dosage , Secondary Metabolism
3.
PLoS One ; 16(7): e0254944, 2021.
Article in English | MEDLINE | ID: mdl-34297759

ABSTRACT

Development of biotherapeutics is hampered by the inherent risk of immunogenicity, which requires extensive clinical assessment and possible re-engineering efforts for mitigation. The focus in the pre-clinical phase is to determine the likelihood of developing treatment-emergent anti-drug antibodies (TE-ADA) and presence of pre-existing ADA in drug-naïve individuals as risk-profiling strategies. Pre-existing ADAs are routinely identified during clinical immunogenicity assessment, but their origin and impact on drug safety and efficacy have not been fully elucidated. One specific class of pre-existing ADAs has been described, which targets neoepitopes of antibody fragments, including Fabs, VH, or VHH domains in isolation from their IgG context. With the increasing number of antibody fragments and other small binding scaffolds entering the clinic, a widely applicable method to mitigate pre-existing reactivity against these molecules is desirable. Here is described a structure-based engineering approach to abrogate pre-existing ADA reactivity to the C-terminal neoepitope of VH(H)s. On the basis of 3D structures, small modifications applicable to any VH(H) are devised that would not impact developability or antigen binding. In-silico B cell epitope mapping algorithms were used to rank the modified VHH variants by antigenicity; however, the limited discriminating capacity of the computational methods prompted an experimental evaluation of the engineered molecules. The results identified numerous modifications capable of reducing pre-existing ADA binding. The most efficient consisted of the addition of two proline residues at the VHH C-terminus, which led to no detectable pre-existing ADA reactivity while maintaining favorable developability characteristics. The method described, and the modifications identified thereby, may provide a broadly applicable solution to mitigate immunogenicity risk of antibody-fragments in the clinic and increase safety and efficacy of this promising new class of biotherapeutics.


Subject(s)
Biological Factors/immunology , Molecular Docking Simulation , Single-Domain Antibodies/chemistry , B-Lymphocytes/immunology , Biological Factors/chemistry , Epitopes/chemistry , Epitopes/immunology , Humans , Protein Binding , Single-Domain Antibodies/immunology
4.
Immunol Invest ; 50(7): 833-856, 2021 Oct.
Article in English | MEDLINE | ID: mdl-33941025

ABSTRACT

Vaccines are an essential part of a preventative healthcare strategy. However, response to vaccines may be less predictable in immunocompromised people. While outcomes for individuals with autoimmune and autoinflammatory diseases have dramatically improved with treatment using immunomodulating and biologic agents, infections have caused significant morbidity in these people today often more than due to their underlying diseases. Immune-based biologic therapies contribute to these infectious complications. This review addresses anti-viral vaccines, their effectiveness and safety in patients treated with approved biologic agents and immune targeted therapy with a focus on vaccines against influenza, human papillomavirus, hepatitis B virus and varicella zoster virus. Preliminary information regarding SARS-CoV-2 anti-viral vaccines is addressed. Additionally, we present recommendations regarding the safe use of vaccines in immunocompromised individuals with the goal to enhance awareness of the safety and efficacy of these anti-viral vaccines in these high-risk populations.


Subject(s)
Antiviral Agents/immunology , Biological Factors/immunology , Hereditary Autoinflammatory Diseases/immunology , Immunologic Factors/immunology , Inflammation/immunology , Virus Diseases/immunology , Viruses/immunology , Hereditary Autoinflammatory Diseases/virology , Humans , Inflammation/virology , Virus Diseases/virology
5.
Pharmacol Res ; 167: 105593, 2021 05.
Article in English | MEDLINE | ID: mdl-33826948

ABSTRACT

Behçet disease (BD) is a complex, multi-systemic inflammatory condition mainly hallmarked by oral and genital ulcers which can also affect the vessels, gastrointestinal tract, central nervous system and even the axial skeleton. Without a clear classification among autoimmune or autoinflammatory conditions, BD has been recently classified as a MHC-I-opathy. BD aetiology is still obscure, but it is thought that certain microorganisms can elicit an aberrant adaptive immune response in the presence of a permissive genetic background. Altered T-cell homeostasis, mostly Th1/Th17 expansion and Treg impairment, could lead to an overactivation of the innate immunity, which underlies tissue damage and thus, signs and symptoms. Immunosuppression and/or immunomodulation are central to the BD management. A complex armamentarium ranging from classical synthetic disease-modifying antirrheumatic drugs to new-era biologic agents or small molecules is available in BD, with different therapeutic outcomes depending on disease manifestations. However, the precise disease mechanisms that underlie BD symptoms are not fully deciphered, which may limit their therapeutic potential and add a significant layer of complexity to the treatment decision-making process. The aim of the present review is to provide an exhaustive overview of the latest breakthroughs in BD pathogenesis and therapeutic options.


Subject(s)
Behcet Syndrome/drug therapy , Biological Factors/therapeutic use , Immunity, Innate/drug effects , Immunomodulation/drug effects , T-Lymphocytes, Regulatory/drug effects , Animals , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/pharmacology , Antibodies, Monoclonal/therapeutic use , Behcet Syndrome/immunology , Biological Factors/immunology , Biological Factors/pharmacology , Humans , Immunity, Innate/immunology , Immunomodulation/immunology , T-Lymphocytes, Regulatory/immunology
6.
Nat Rev Rheumatol ; 17(2): 81-97, 2021 02.
Article in English | MEDLINE | ID: mdl-33318665

ABSTRACT

Biologic agents have become a core component of therapeutic strategies for many inflammatory rheumatic diseases. However, perhaps reflecting the specificity and generally high affinity of biologic agents, these therapeutics have been used by rheumatologists with less consideration of their pharmacokinetics than that of conventional synthetic DMARDs. Immunogenicity was recognized as a potential limitation to the use of biologic agents at an early stage in their development, although regulatory guidance was relatively limited and assays to measure immunogenicity were less sophisticated than today. The advent of biosimilars has sparked a renewed interest in immunogenicity that has resulted in the development of increasingly sensitive assays, an enhanced appreciation of the pharmacokinetic consequences of immunogenicity and the development of comprehensive and specific guidance from regulatory authorities. As a result, rheumatologists have a greatly improved understanding of the field in general, including the factors responsible for immunogenicity, its potential clinical consequences and the implications for everyday treatment. In some specialties, immunogenicity testing is becoming a part of routine clinical management, but definitive evidence of its cost-effectiveness in rheumatology is awaited.


Subject(s)
Adaptive Immunity/drug effects , Biological Factors/pharmacokinetics , Rheumatic Diseases/drug therapy , Rheumatology/standards , Adaptive Immunity/immunology , Antirheumatic Agents/administration & dosage , Antirheumatic Agents/therapeutic use , Biological Factors/immunology , Biological Factors/therapeutic use , Biosimilar Pharmaceuticals/pharmacokinetics , Biosimilar Pharmaceuticals/therapeutic use , Cost-Benefit Analysis , Humans , Rheumatic Diseases/immunology , Rheumatologists/statistics & numerical data , Rheumatology/economics
7.
Expert Rev Clin Pharmacol ; 14(1): 47-53, 2021 Jan.
Article in English | MEDLINE | ID: mdl-32432941

ABSTRACT

INTRODUCTION: Biological agents (BAs) target molecules involved in disease mechanisms and have modified the natural history of several immune-mediated disorders. All BAs are immunogenic, resulting in the formation of antidrug antibodies (ADAs), which can neutralize drug activity leading to loss of response and potential relapse, or serious adverse events such as infusion hypersensitivity reactions. The production of ADAs is the result of a specific adaptive immune response in which T and B cells are involved. AREAS COVERED: Factors conditioning the immunogenicity of BAs, including drug-, treatment- and patient-related factors are currently the subject of many studies. Among them, a lot of attention is dedicated to define the impact of BAs structure, the effect of targeting (soluble or membrane) molecules, the impact of interruption of therapy as well as the role of genetic (HLA and non-HLA) predisposing factors and disease activity. EXPERT OPINION: Knowledge of factors capable of influencing the immunogenicity of BAs may help to understand, in a predictive manner and at the single patient level, the presence of risk factors influencing the production of ADAs and their impact on clinical outcomes.


Subject(s)
Antibody Formation/immunology , Biological Factors/adverse effects , Animals , B-Lymphocytes/immunology , Biological Factors/administration & dosage , Biological Factors/immunology , Humans , Immunity/immunology , Risk Factors , T-Lymphocytes/immunology
8.
Ann Lab Med ; 40(2): 101-113, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31650726

ABSTRACT

Biological drugs, such as proteins and immunogens, are increasingly used to treat various diseases, including tumors and autoimmune diseases, and biological molecules have almost completely replaced synthetic drugs in rheumatology. Although biological treatments such as anti-tumor necrosis factor (TNF) drugs seem to be quite safe, they cause some undesirable effects, such as the onset of infections due to weakening of the immune system. Given the biological nature of these drugs, they might be recognized as extraneous; this would induce an immune reaction that neutralizes their effectiveness or lead to more serious consequences. Laboratories play a pivotal role in appropriate therapeutic management. The aim of this review was to underline the production of anti-drug antibodies during treatment with biological drugs and highlight the role of laboratories in ensuring appropriate use of these drugs.


Subject(s)
Biological Factors/blood , Drug Monitoring , Adalimumab/blood , Adalimumab/immunology , Adalimumab/therapeutic use , Antibodies, Monoclonal/blood , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Arthritis, Rheumatoid/drug therapy , Biological Factors/immunology , Biological Factors/therapeutic use , Biosimilar Pharmaceuticals/blood , Biosimilar Pharmaceuticals/therapeutic use , Humans
9.
Rheumatology (Oxford) ; 58(10): 1839-1849, 2019 10 01.
Article in English | MEDLINE | ID: mdl-30809664

ABSTRACT

OBJECTIVE: The clinical impact of anti-drug antibodies (ADAbs) in paediatric patients with JIA remains unknown. This systematic review and meta-analysis aimed to summarize the prevalence of ADAbs in JIA studies; investigate the effect of ADAbs on treatment efficacy and adverse events; and explore the effect of immunosuppressive therapy on antibody formation. METHODS: PubMed, Embase and the Cochrane Library were systematically searched to identify relevant clinical trials and observational studies that reported prevalence of ADAbs. Studies were systematically reviewed in accordance with the Preferred Reporting Items for Systematic reviews and Meta-Analyses and appropriate proportional and pairwise meta-analyses were performed. RESULTS: A total of 5183 references were screened; 28 articles, involving 26 studies and 2354 JIA patients, met eligibility criteria. Prevalence of ADAbs ranged from 0% to 82% across nine biologic agents. Overall pooled prevalence of ADAbs was 16.9% (95% CI, 9.5, 25.9). Qualitative analysis of included studies indicated that antibodies to infliximab, adalimumab, anakinra and tocilizumab were associated with treatment failure and/or hypersensitivity reactions. Concomitant MTX uniformly reduced the risk of antibody formation during adalimumab treatment (risk ratio 0.33; 95% CI 0.21, 0.52). CONCLUSION: The association of ADAbs with treatment failure and hypersensitivity reactions indicates their clinical relevance in paediatric patients with JIA. Based on our findings, we recommend a preliminary course of action regarding immunogenicity of biologic agents in patients with JIA. Further strategies to predict, prevent, detect and manage immunogenicity could optimize treatment outcomes and personalize treatment with biologic therapies.


Subject(s)
Antibody Formation , Antirheumatic Agents/immunology , Arthritis, Juvenile/drug therapy , Arthritis, Juvenile/immunology , Biological Factors/immunology , Adalimumab/immunology , Antibodies, Monoclonal, Humanized/immunology , Child , Clinical Trials as Topic , Humans , Infliximab/immunology , Interleukin 1 Receptor Antagonist Protein/immunology , Methotrexate/immunology , Observational Studies as Topic
10.
Clin Pharmacol Ther ; 105(4): 970-978, 2019 04.
Article in English | MEDLINE | ID: mdl-30372517

ABSTRACT

A mathematical antidrug antibody (ADA) model was developed to quantitatively assess immunogenicity for therapeutic proteins. The ADA model was built with antibody titer data in subjects from 10 clinical trials. The time course of the antibody titers was quantitatively characterized with a two-component semimechanistic model describing the double peaks of ADA titers. The relationship between antibody titer and incidence was also explored. The ADA incidences in subjects from 12 clinical trials were used for internal and external validations. The ADA titers reasonably predicted the incidence of antibody. The model-predicted elimination rate constant for antibody titer was 14.1 × 10-3  day-1 and 8.1 × 10-3  day-1 in healthy subjects and patients, respectively. This research provided a useful tool to quantitatively evaluate immunogenicity and its impact for therapeutic proteins.


Subject(s)
Antibodies, Monoclonal/immunology , Proteins/immunology , Adult , Biological Factors/immunology , Clinical Trials as Topic , Female , Healthy Volunteers , Humans , Male , Middle Aged , Models, Theoretical
11.
J Int Med Res ; 46(10): 4061-4070, 2018 Oct.
Article in English | MEDLINE | ID: mdl-30152254

ABSTRACT

Objective To investigate the relationship between inflammatory factors, oxidative stress and type 1 deiodinase (DIO-1) concentration in patients with chronic renal failure (CRF) with or without euthyroid sick syndrome (ESS). Methods This study recruited patients with CRF and divided them into two groups: group 1 had low free triiodothyronine (FT3) levels; and group 2 had normal FT3 levels. Group 3 consisted of healthy volunteers. Serum levels of interleukin (IL)-6, IL-1ß, tumour necrosis factor (TNF)-α, 8-isoprostane and DIO-1 were measured using enzyme-linked immunosorbent assays. Multiple regression analysis was used to analyse correlations between parameters. Results Sixty patients were enrolled into each group and the groups were comparable in terms of vital signs, white blood cell count, free thyroxine and thyroid stimulating hormone concentrations. The serum DIO-1 concentration was significantly higher in group 2 than in groups 1 and 3. Multivariate regression analysis revealed that the DIO-1 concentration was inversely correlated with the TNF-α concentration. Conclusions Patients with CRF without ESS showed higher concentrations of DIO-1 than patients with ESS. The DIO-1 concentration was inversely correlated with the TNF-α concentration, which might indicate that the inflammatory response was milder in the patients with CRF without ESS than in those with ESS.


Subject(s)
DNA-Binding Proteins/blood , Euthyroid Sick Syndromes/immunology , Inflammation/immunology , Kidney Failure, Chronic/immunology , Oxidative Stress/immunology , Aged , Biological Factors/blood , Biological Factors/immunology , Cytokines/blood , Cytokines/immunology , DNA-Binding Proteins/immunology , Euthyroid Sick Syndromes/blood , Female , Humans , Inflammation/blood , Kidney Failure, Chronic/blood , Male , Middle Aged , Triiodothyronine/blood , Triiodothyronine/immunology
12.
Expert Rev Clin Immunol ; 14(6): 513-523, 2018 06.
Article in English | MEDLINE | ID: mdl-29683362

ABSTRACT

INTRODUCTION: Antibodies or fusion proteins termed biologics allow the targeted therapy of diseases. Many of these agents have proven superior efficacy and safety to conventional therapies, and subsequently revolutionized the management of numerous chronic diseases. Repetitive administration of these protein-based therapeutics to immunocompetent patients elicit immune responses in the form of Anti Drug Antibodies (ADAs), which in turn impact their pharmacological properties and may trigger adverse events. Areas covered: Structural characteristics determining the immunogenicity of biologics are reviewed along with strategies to minimize it. Next, the different types of treatment-emerging ADAs, their potential clinical implications, and assays to detect them are addressed. Emphasis is put on the review of data on the immunogenicity of different types of biologics across numerous indications. Finally, practical considerations are discussed on how to manage patients with issues around the immunogenicity of their biologic treatment. Expert commentary: Immunogenicity is a clinically relevant criterion when selecting a biologic. Besides intrinsic properties of the agent (namely its structure), its respective mode of action, dosing regimen, comedication, and the indication treated must be considered. ADA detection assays need to be standardized to improve comparability of available data and to allow clinical decision-making.


Subject(s)
Antibody Formation/immunology , Biological Factors/immunology , Humans
13.
Front Immunol ; 9: 233, 2018.
Article in English | MEDLINE | ID: mdl-29497421

ABSTRACT

T regulatory cells, a specialized subset of T cells, are key players in modulating antigen (Ag)-specific immune responses in vivo. Inducible T regulatory type 1 (Tr1) cells are characterized by the co-expression of CD49b and lymphocyte-activation gene 3 (LAG-3) and the ability to secrete IL-10, TGF-ß, and granzyme (Gz) B, in the absence of IL-4 and IL-17. The chief mechanisms by which Tr1 cells control immune responses are secretion of IL-10 and TGF-ß and killing of myeloid cells via GzB. Tr1 cells, first described in peripheral blood of patients who developed tolerance after HLA-mismatched fetal liver hematopoietic stem cell transplantation, have been proven to modulate inflammatory and effector T cell responses in several immune-mediated diseases. The possibility to generate and expand Tr1 cells in vitro in an Ag-specific manner has led to their clinical use as cell therapy in patients. Clinical grade protocols to generate or to enrich and expand Tr1 cell medicinal products have been established. Proof-of-concept clinical trials with Tr1 cell products have demonstrated the safety and the feasibility of this approach and indicated some clinical benefit. In the present review, we provide an overview on protocols established to induce/expand Tr1 cells in vitro for clinical application and on results obtained in Tr1 cell-based clinical trials. Moreover, we will discuss a recently developed protocol to efficient convert human CD4+ T cells into a homogeneous population of Tr1-like cells by lentiviral vector-mediated IL-10 gene transfer.


Subject(s)
Cell Engineering/methods , Immune Tolerance , Immunotherapy/methods , Interleukin-10/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigens, CD/immunology , Antigens, CD/metabolism , Biological Factors/immunology , Biological Factors/metabolism , Biological Factors/therapeutic use , Cell Culture Techniques/methods , Cell Engineering/trends , Cell Line , Cell Transplantation/methods , Clinical Trials as Topic , Disease Models, Animal , Gene Transfer Techniques , Humans , Integrin alpha2/immunology , Integrin alpha2/metabolism , Interleukin-10/genetics , Interleukin-10/metabolism , Mice , T-Lymphocytes, Regulatory/transplantation , Lymphocyte Activation Gene 3 Protein
14.
Expert Opin Biol Ther ; 18(5): 575-584, 2018 05.
Article in English | MEDLINE | ID: mdl-29533116

ABSTRACT

INTRODUCTION: Biologic agents have demonstrated efficacy in treating patients with psoriatic arthritis (PsA). Biologic agents also have an intrinsic capacity to induce an immune response in patients that could result in unwanted adverse events and/or treatment failure. AREAS COVERED: In this systematic literature review, the authors document the incidence of immune responses, primarily anti-drug antibodies (ADA), to the biologic therapeutic agents currently in clinical practice for the treatment of PsA. The authors discuss the importance of these responses with respect to clinical practice. EXPERT OPINION: Our evaluation of the published literature shows that the immune responses to the various biologic therapeutic agents currently being used to treat PsA are similar to those observed for these agents in other rheumatic diseases. Moreover, similar to observations in other rheumatic diseases, the incidence of ADA formation to biologic agents in patients with PsA is often decreased when patients are given concomitant treatment with disease-modifying anti-rheumatic drugs. These data strongly suggest that the immune response is a characteristic of the biologic agent. Using therapeutic drug monitoring may be an approach to assess the immune response to the agent and to mitigate the potential impact on efficacy and safety, and consequently optimize treatment.


Subject(s)
Antirheumatic Agents/immunology , Antirheumatic Agents/therapeutic use , Arthritis, Psoriatic/drug therapy , Biological Factors/immunology , Biological Factors/therapeutic use , Antibodies, Monoclonal/immunology , Antibodies, Monoclonal/therapeutic use , Arthritis, Psoriatic/immunology , Biological Therapy/adverse effects , Biological Therapy/methods , Humans , Immunogenetic Phenomena
15.
Food Chem Toxicol ; 116(Pt A): 32-41, 2018 Jun.
Article in English | MEDLINE | ID: mdl-29454867

ABSTRACT

Microbial-based cleaning products (MBCPs) contain bacteria and chemical constituents. They are used in consumer applications such as odor reduction, unclogging drains, and surface cleaning. To determine the capacity of a model MBCP to contribute to acute allergic lung inflammation, a two-week repeated exposure regimen was used. Mice were exposed by endotracheal instillation to saline alone, MBCP alone, house dust mites (HDM) alone, or sequentially (i.e., MBCP followed by HDM, HDM followed by MBCP, or HDM + MBCP followed by HDM). Both whole MBCP and acellular MBCP filtrate were investigated, and showed minimal differences in the endpoints examined. HDM exposure caused pulmonary perivascular inflammation, bronchiolar mucous cell metaplasia, elevated bronchoalveolar lavage fluid (BALF) eosinophils, and HDM-specific IgG1. For MBCP, notable changes were associated with sequential exposures. MBCP/HDM caused elevated TH2 cytokines in BALF, and elevated neutrophils, eosinophils and IL-5 in peripheral blood. Co-administration of MBCP and HDM followed by HDM resulted in elevated blood and BALF eosinophils and HDM-specific IgE and IgG1. These results demonstrated that acellular MBCP filtrate, and not bacteria within MBCPs, potentiated the acute allergic inflammation to HDM. This methodology could be extended to investigate chronic allergic inflammation and inflammatory potential of other MBCPs and biotechnology products with complex compositions.


Subject(s)
Biological Factors/immunology , Detergents/adverse effects , Pneumonia/immunology , Pyroglyphidae/immunology , Animals , Biological Factors/adverse effects , Biological Factors/analysis , Bronchoalveolar Lavage Fluid/immunology , Detergents/chemistry , Disease Models, Animal , Eosinophils/immunology , Female , Humans , Immunoglobulin E/immunology , Lung/immunology , Mice , Mice, Inbred BALB C , Pneumonia/etiology , Quality Control
16.
Curr Protein Pept Sci ; 19(10): 958-971, 2018.
Article in English | MEDLINE | ID: mdl-28847291

ABSTRACT

Currently it is well known that all biological drugs, including those with a fully human structure, are capable of inducing a host immune response known as immunogenicity [1]. The presence of ADAs can condition the drug´s level and action, thus modifying the therapeutic effect and even the safety profile by its mechanism of action - neutralizing or non-neutralizing - and / or an increase in its clearance. Immunogenicity is a dynamic factor to be taken into account in biological therapy, especially in long-term treatments, and as a relevant aspect in the assessment of secondary response loss [2]. With the above, not only the knowledge but also the management of the immunogenicity of the different biological treatments, represent a useful instrument for optimization of the strategies of use for each drug, and in the design of predictive models of response, which finally permits a significant improvement in the efficacy and safety profile, aiming to a personalization of the therapies, especially in patients with autoimmune diseases, genetic disorders and cancer [3]. This review summarizes the events of immunogenicity that produce the biological drug, the factor that influence to immunogenicity and the assessment of immunogenicity.


Subject(s)
Biological Factors/immunology , Proteins/immunology , Adaptive Immunity , Antibodies/blood , Antibodies/immunology , Autoimmune Diseases/drug therapy , Autoimmune Diseases/immunology , Biological Factors/pharmacology , Biological Factors/therapeutic use , Biosimilar Pharmaceuticals/pharmacology , Biosimilar Pharmaceuticals/therapeutic use , Genetic Diseases, Inborn/drug therapy , Genetic Diseases, Inborn/immunology , Humans , Immunity, Active , Immunity, Humoral , Neoplasms/drug therapy , Neoplasms/immunology , Peptides/immunology , Peptides/pharmacology , Peptides/therapeutic use , Proteins/pharmacology , Proteins/therapeutic use
17.
Expert Opin Biol Ther ; 18(3): 309-315, 2018 03.
Article in English | MEDLINE | ID: mdl-29186988

ABSTRACT

INTRODUCTION: Since 2006, biosimilars have been available in several countries worldwide, thus allowing for potential savings in pharmaceutical expenditure. However, there have been numerous debates about the interchangeability of biosimilars and reference products based on concerns of immunogenicity by switching between biological products, which may cause lack of effect and toxicity. AREAS COVERED: The authors provide the reader with an overview of the different positions of regulatory authorities on the interchangeability and automatic substitution of biosimilars and reference products. Presently, the FDA allows automatic substitution without prescriber intervention if the biosimilar is interchangeable with reference products, while the European Medicines Agency delegate to each single EU member state. EXPERT OPINION: Different approaches in defining interchangeability and automatic substitution call for harmonization to increase confidence of healthcare professionals and patients about the clinical impact of switching. Networks of electronic healthcare records and administrative databases, potentially linkable to clinical charts and registries may rapidly assess frequency and benefit-risk profile of different switching patterns in routine care at different levels, thus integrating and strengthening pre-marketing evidence.


Subject(s)
Autoimmune Diseases/drug therapy , Biosimilar Pharmaceuticals/therapeutic use , Neoplasms/drug therapy , Antibodies, Anti-Idiotypic/blood , Autoimmune Diseases/pathology , Biological Factors/immunology , Biological Factors/pharmacokinetics , Biological Factors/therapeutic use , Biosimilar Pharmaceuticals/economics , Biosimilar Pharmaceuticals/pharmacokinetics , Humans , Neoplasms/pathology , Product Surveillance, Postmarketing
18.
Rheumatology (Oxford) ; 57(2): 246-254, 2018 Feb 01.
Article in English | MEDLINE | ID: mdl-28541488

ABSTRACT

In this review we discuss the divergent role of two closely related cytokines, IL-12 and IL-23, in shaping immune responses. In light of current therapeutic developments using biologic agents to block these two pathways, a better understanding of the immunological function of these cytokines is pivotal.


Subject(s)
Inflammatory Bowel Diseases/immunology , Interleukin-12/immunology , Interleukin-23/immunology , Psoriasis/immunology , T-Lymphocytes/immunology , Biological Factors/immunology , Biological Factors/therapeutic use , Chronic Disease , Humans , Immunologic Factors/immunology , Immunologic Factors/therapeutic use , Inflammation/immunology , Inflammatory Bowel Diseases/therapy , Interleukin-12/antagonists & inhibitors , Interleukin-12/deficiency , Interleukin-23/antagonists & inhibitors , Lymphocyte Activation/immunology , Psoriasis/therapy
19.
Bioanalysis ; 9(22): 1733-1737, 2017 Nov.
Article in English | MEDLINE | ID: mdl-29120228

ABSTRACT

The 8th Japan Bioanalysis Forum symposium, the Tower Hall Funabori, Tokyo, Japan, 8-9 February 2017 The 8th Japan Bioanalysis Forum (JBF) symposium was successfully held between 8 and 9 February 2017 at the Tower Hall Funabori, Tokyo, Japan. In total, 24 speakers from Japan, USA and Europe gave presentations regarding the immunogenicity of biopharmaceuticals, ICH S3A Q&A microsampling, ICH M10 bioanalytical method validation, large molecule analysis through LC-MS, auditing activities for bioanalysis and biomarker bioanalysis. Achievements regarding eight diverse themes were also shared by Japan Bioanalysis Forum discussion groups. Over 300 scientists from regulatory agencies, industry and academia actively took part in discussions during the symposium. This article provides the highlights of all the topics discussed in this symposium.


Subject(s)
Biomarkers/analysis , Biological Factors/immunology , Chromatography, High Pressure Liquid , Government Regulation , Japan , Mass Spectrometry
20.
Allergy Asthma Proc ; 38(6): 409-418, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29046192

ABSTRACT

Hypersensitivity reactions (HSR) to targeted biologic agents present as immediate reactions during infusion or as delayed reactions (after one or more exposures). The new classification includes phenotypes, endotypes, and biomarkers. Phenotypes include immediate type I (immunoglobulin E [IgE] or non-IgE mediated), cytokine release, mixed (type I/cytokine), and immune complexes type III (IgG mediated) reactions as well as delayed type IV reactions. Endotypes include IgE or non-IgE mediated mast cells/basophils activation with elevated serum tryptase and T cells values as well as macrophages, which lead to cytokine production, e.g., interleukin 6. A skin test is a valuable tool in evaluating HSRs to biologics; however, its predictive value depends on the type of reaction and the monoclonal antibody. Desensitization is a new approach to safely reintroduce biologics when they are first-line therapies, and it is available for immediate and delayed reactions. Research is needed to further understand the mechanisms of reactions to monoclonal antibodies and their management.


Subject(s)
Biological Factors/adverse effects , Desensitization, Immunologic/methods , Drug Hypersensitivity/therapy , Biological Factors/immunology , Biological Factors/therapeutic use , Biomarkers , Humans , Phenotype
SELECTION OF CITATIONS
SEARCH DETAIL
...