101
|
Muthas D, Reznichenko A, Balendran CA, Böttcher G, Clausen IG, Kärrman Mårdh C, Ottosson T, Uddin M, MacDonald TT, Danese S, Berner Hansen M. Neutrophils in ulcerative colitis: a review of selected biomarkers and their potential therapeutic implications. Scand J Gastroenterol 2017; 52:125-135. [PMID: 27610713 DOI: 10.1080/00365521.2016.1235224] [Citation(s) in RCA: 115] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
OBJECTIVES This review article describes the role of neutrophils in mucosal injury and the resulting crypt abscesses characteristic of ulcerative colitis. We also review selected biomarkers for monitoring neutrophil presence and activity in the mucosa as well as their potential as therapeutic targets. MATERIAL We have collated and selectively reviewed data on the most prominent well-established and emerging neutrophil-related biomarkers and potential therapeutic targets (calprotectin, lactoferrin, CXCR1, CXCR2, MMP-9, NGAL, elafin, HNE, pANCAs, MPO, CD16, CD177, CD64, HNPs, SLPI and PTX3) in ulcerative colitis. RESULTS Systemic and intestinal neutrophil activity increases substantially in active ulcerative colitis, driving tissue damage and extra-intestinal manifestations. Calprotectin is a robust neutrophil and disease biomarker, and a few neutrophil-related targets are being clinically explored as therapeutic targets. CONCLUSION We propose that targeting neutrophils and their inflammatory mediators per se is an opportunity that should be explored to identify new effective medical therapies. The overall clinical goal for neutrophil-targeted therapy will be to modulate, but not completely silence, neutrophil activity, thereby abolishing the destructive inflammation with associated acute and chronic tissue damage without compromising host-defense.
Collapse
Affiliation(s)
- Daniel Muthas
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Anna Reznichenko
- b Department of Cardiovascular and Metabolic Diseases , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Clare A Balendran
- c Department of Personalised HealthCare & Biomarkers , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Gerhard Böttcher
- d Department of Drug Safety and Metabolism , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Ib Groth Clausen
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Carina Kärrman Mårdh
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Tomas Ottosson
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Mohib Uddin
- c Department of Personalised HealthCare & Biomarkers , AstraZeneca R&D Gothenburg , Mölndal , Sweden
| | - Thomas T MacDonald
- e Blizard Institute, Barts and the London School of Medicine and Dentistry, QMUL , London , UK
| | - Silvio Danese
- f Department of Gastroenterology , IBD Center, Humanitas Research Hospital , Milan , Italy
| | - Mark Berner Hansen
- a Department of Respiratory , Inflammation and Autoimmunity, AstraZeneca R&D Gothenburg , Mölndal , Sweden.,g Digestive Disease Center K, Bispebjerg Hospital, University of Copenhagen , Copenhagen , Denmark
| |
Collapse
|
102
|
Chung SI, Horton JA, Ramalingam TR, White AO, Chung EJ, Hudak KE, Scroggins BT, Arron JR, Wynn TA, Citrin DE. IL-13 is a therapeutic target in radiation lung injury. Sci Rep 2016; 6:39714. [PMID: 28004808 PMCID: PMC5177927 DOI: 10.1038/srep39714] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2016] [Accepted: 11/25/2016] [Indexed: 01/08/2023] Open
Abstract
Pulmonary fibrosis is a potentially lethal late adverse event of thoracic irradiation. Prior research indicates that unrestrained TGF-β1 and/or type 2 cytokine-driven immune responses promote fibrosis following radiation injury, but the full spectrum of factors governing this pathology remains unclear. Interleukin 13 (IL-13) is a key factor in fibrotic disease associated with helminth infection, but it is unclear whether it plays a similar role in radiation-induced lung fibrosis. Using a mouse model, we tested the hypothesis that IL-13 drives the progression of radiation-induced pulmonary fibrosis. Irradiated lungs from wild-type c57BL/6NcR mice accumulated alternatively-activated macrophages, displayed elevated levels of IL-13, and extensive fibrosis, whereas IL-13 deficient mice were resistant to these changes. Furthermore, plasma from irradiated wild-type mice showed a transient increase in the IL-13 saturated fraction of the circulating decoy receptor IL-13Rα2. Finally, we determined that therapeutic neutralization of IL-13, during the period of IL-13Rα2 saturation was sufficient to protect mice from lung fibrosis. Taken together, our results demonstrate that IL-13 is a major regulator of radiation-induced lung injury and demonstrates that strategies focusing on IL-13 may be useful in screening for timely delivery of anti-IL-13 therapeutics.
Collapse
Affiliation(s)
- Su I Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Jason A Horton
- Musculoskeletal Science Research Center, Dept. of Orthopedic Surgery, Upstate Medical University, Syracuse, New York, USA
| | | | - Ayla O White
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Eun Joo Chung
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Kathryn E Hudak
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Bradley T Scroggins
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| | - Joseph R Arron
- Biomarker Discovery OMNI, Genentech, Inc. MS 231c, 1 DNA way, San Francisco, CA 94080 USA
| | - Thomas A Wynn
- Laboratory of Parasitic Diseases, National Institute of Allergy and Infectious Diseases, 4 Memorial Drive, Room 211C, Bethesda, MD 20892-0425, USA
| | - Deborah E Citrin
- Radiation Oncology Branch, Center for Cancer Research, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
103
|
Bilsborough J, Targan SR, Snapper SB. Therapeutic Targets in Inflammatory Bowel Disease: Current and Future. ACTA ACUST UNITED AC 2016. [DOI: 10.1038/ajgsup.2016.18] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
104
|
Boyapati RK, Kalla R, Satsangi J, Ho GT. Biomarkers in Search of Precision Medicine in IBD. Am J Gastroenterol 2016; 111:1682-1690. [PMID: 27670602 DOI: 10.1038/ajg.2016.441] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Accepted: 08/02/2016] [Indexed: 12/11/2022]
Abstract
The completion of the human genome project in 2003 represented a major scientific landmark, ushering in a new era with hopes and expectations of fresh insights into disease mechanisms and treatments. In inflammatory bowel disease (IBD), many important discoveries soon followed, notably the identification of >200 genetic susceptibility loci and characterization of the gut microbiome. As "big data", driven by advances in technology, becomes increasingly available and affordable, individuals with IBD and clinicians alike yearn for tangible outcomes from the promise of "precision medicine"-precise diagnosis, monitoring, and treatment. Here, we provide a commentary on the prospects and challenges of precision medicine and biomarkers in IBD. We focus on the three key areas where precision IBD will have the most impact: (1) disease susceptibility, activity, and behavior; (2) prediction of drug response and adverse effects; and (3) identification of subphenotypic mechanisms to facilitate drug discovery and selection of new treatments in IBD.
Collapse
Affiliation(s)
- Ray K Boyapati
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Department of Gastroenterology, Monash Health, Clayton, Victoria, Australia
| | - Rahul Kalla
- Gastrointestinal Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - Jack Satsangi
- Gastrointestinal Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| | - Gwo-Tzer Ho
- MRC Centre for Inflammation Research, Queens Medical Research Institute, University of Edinburgh, Edinburgh, UK.,Gastrointestinal Unit, Institute of Genetics and Molecular Medicine, Western General Hospital, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
105
|
Föhlinger M, Palamides P, Mansmann U, Beigel F, Siebeck M, Gropp R. Immunological profiling of patients with ulcerative colitis leads to identification of two inflammatory conditions and CD1a as a disease marker. J Transl Med 2016; 14:310. [PMID: 27809916 PMCID: PMC5094062 DOI: 10.1186/s12967-016-1048-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2016] [Accepted: 10/03/2016] [Indexed: 01/09/2023] Open
Abstract
Background Conventional approaches to understand mechanisms underlying the development of pathological manifestations in ulcerative colitis (UC) mostly rely on identification of certain cell types and cytokines followed by verification of their roles in vitro and in vivo. In light of the highly dynamic processes in UC, requiring the cross talk of immune cells, epithelial-, endothelial-, muscle cells and fibrocytes, this approach might neglect temporal and spatial connectivity of individually differing inflammatory responses. Methods We undertook a more holistic approach whereby we designed a flow cytometric analysis- and ELISA panel and determined the immunological profiles of UC patients in comparison to Non UC donors. This panel consisted of B-cells, T-cells, macrophages, monocytes, NK- and NK T-cells and subtypes thereof, the cytokines TGFß1 and HGF, the chemokine TARC and periostin. Blood was collected from 41 UC patients and 30 non-UC donors. Isolated PBMC were subjected to flow cytometric analysis and sera were analyzed by ELISA. Data were analysed by cluster- and correlation analysis. To corroborate that the identified cells reflected the inflammatory condition in the colon of UC patients, leucocytes were isolated from colons of UC patients and subjected to the same flow cytometric analysis. Results Immunological profiling followed by cluster- and correlation analysis led to the identification of two inflammatory conditions: An ‘acute’ condition characterized by adaptive immune cells as plasma cells, TSLPR expressing CD11b+ macrophages, CD64 and CCR2 expressing CD14+ monocytes, HGF and TARC and a ‘remodeling’ condition signified by NK T-cells and TLSPR expressing CD14+ monocytes, TGFß1 and periostin. ROC analysis identified TARC and TGFß1 as biological markers with high potential to discriminate between these two conditions (Δ = −6687.72 ng/ml; p = 1E−04; AUC = 0.87). In addition, CD1a+ CD11b+ macrophages (Δ = 17.73% CD1a+ CD11b+; p = 5E−04; AUC = 0.86) and CD1a+ CD14+ monocytes (Δ = 20.35; p = 0.02, AUC = 0.75) were identified as markers with high potential to discriminate between UC and Non UC donors. CD1a+ CD11b+ macrophages and NK T-cells were found to be significantly increased in inflamed colons of UC patients as compared to non-UC control samples (p = 0.02). Conclusions Immunological profiling of UC patients might improve our understanding of the pathology underlying individual manifestations and phases of the disease. This might lead to the development of novel diagnostics and therapeutic interventions adapted to individual needs and different phases of the disease. In addition, it might result in stratification of patients for clinical trials. Electronic supplementary material The online version of this article (doi:10.1186/s12967-016-1048-9) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- M Föhlinger
- Department of General Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Nussbaumstr. 20, 80336, Munich, Germany
| | - P Palamides
- Institute of Molecular Animal Breeding and Biotechnology, and Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, 81377, Munich, Germany
| | - U Mansmann
- Institute for Medical Informatics, Biometry and Epidemiology, LMU Munich, Marchioninistr. 15, 81377, Munich, Germany
| | - F Beigel
- Department of Medicine II-Grosshadern, Ludwig-Maximilians-University (LMU), Marchioninistr. 15, 81377, Munich, Germany
| | - M Siebeck
- Department of General Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Nussbaumstr. 20, 80336, Munich, Germany
| | - R Gropp
- Department of General Visceral, and Transplantation Surgery, Hospital of the LMU Munich, Nussbaumstr. 20, 80336, Munich, Germany.
| |
Collapse
|
106
|
Inflammatory bowel disease: exploring gut pathophysiology for novel therapeutic targets. Transl Res 2016; 176:38-68. [PMID: 27220087 DOI: 10.1016/j.trsl.2016.04.009] [Citation(s) in RCA: 133] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2015] [Revised: 03/17/2016] [Accepted: 04/28/2016] [Indexed: 12/14/2022]
Abstract
Ulcerative colitis and Crohn's disease are the 2 major phenotypes of inflammatory bowel disease (IBD), which are influenced by a complex interplay of immunological and genetic elements, though the precise etiology still remains unknown. With IBD developing into a globally prevailing disease, there is a need to explore new targets and a thorough understanding of the pathophysiological differences between the healthy and diseased gut could unearth new therapeutic opportunities. In this review, we provide an overview of the major aspects of IBD pathogenesis and thereafter present a comprehensive analysis of the gut pathophysiology leading to a discussion on some of the most promising targets and biologic therapies currently being explored. These include various gut proteins (CXCL-10, GATA-3, NKG2D, CD98, microRNAs), immune cells recruited to the gut (mast cells, eosinophils, toll-like receptors 2, 4), dysregulated proinflammatory cytokines (interleukin-6, -13, -18, -21), and commensal microbiota (probiotics and fecal microbiota transplantation). We also evaluate some of the emerging nonconventional therapies being explored in IBD treatment focusing on the latest developments in stem cell research, oral targeting of the gut-associated lymphoid tissue, novel anti-inflammatory signaling pathway targeting, adenosine deaminase inhibition, and the beneficial effects of antioxidant and nutraceutical therapies. In addition, we highlight the growth of biologics and their targets in IBD by providing information on the preclinical and clinical development of over 60 biopharmaceuticals representing the state of the art in ulcerative colitis and Crohn's disease drug development.
Collapse
|
107
|
Wang X, Zhu Y, Zhang M, Wang H, Jiang Y, Gao P. Ulcerative Colitis Is Characterized by a Decrease in Regulatory B Cells. J Crohns Colitis 2016; 10:1212-23. [PMID: 26980839 DOI: 10.1093/ecco-jcc/jjw074] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2015] [Accepted: 03/11/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND AND AIMS Ulcerative colitis (UC) is characterized by Th2-polarized, B cell responses. However, neither size, composition, nor activity of the Breg compartment in active UC has been evaluated. METHODS Peripheral blood of 25 patients with UC, 5 patients with Crohn's disease (CD) and 15 control subjects was examined for Bregs subsets by flow cytometry. Intestinal tissue of 5 patients with UC, 3 patients with CD and 5 control subjects was also examined for Bregs. The levels of serum interleukin (IL)-10, immunoglobulin (Ig), and C-reactive protein (CRP), and the erythrocyte sedimentation rate (ESR) in individual subjects were measured. Interleukin-10 production in B cells isolated from patients with UC was examined. Potential associations between the values of the different measures were analysed by the Spearman correlation test. RESULTS Compared with controls, the UC patients had significantly reduced frequencies of CD24(high)CD38(high) and CD5(+) Bregs in peripheral blood as well as in intestinal tissues, accompanied by lower serum IL-10 levels. Interleukin-10 production was significantly decreased in stimulated B cells from patients with UC, whereas patient IL-10(+) B cells were found to be enriched in CD24(high)CD38(high) and CD5(+) B cells. However, increased percentages of CD95(+)-exhausted Bregs were encountered in subsets. Furthermore, Mayo clinic scores, CRP, and ESR in UC patients was negatively correlated with the frequency of Bregs and the IL-10 concentration, whereas these parameters were positively correlated with the frequency of CD95(+)-exhausted Bregs and the IgG levels. CONCLUSIONS Active UC is characterized by exhaustion of regulatory control in the B cell compartment.
Collapse
Affiliation(s)
- Xinrui Wang
- Department of Central Laboratory, the First Hospital, Jilin University, Changchun 130021, China
| | - Yonggang Zhu
- Department of Radiation Oncology, China-Japan Union Hospital of Jilin University, Changchun 130021, China
| | - Manli Zhang
- Department of Central Laboratory, the First Hospital, Jilin University, Changchun 130021, China
| | - Hongjuan Wang
- Department of Central Laboratory, the First Hospital, Jilin University, Changchun 130021, China
| | - Yanfang Jiang
- Department of Central Laboratory, the First Hospital, Jilin University, Changchun 130021, China Key Laboratory of Zoonosis Research, Ministry of Education, the First Hospital, Jilin University, Changchun 130021, China Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou 225009, China
| | - Pujun Gao
- Department of Central Laboratory, the First Hospital, Jilin University, Changchun 130021, China
| |
Collapse
|
108
|
Palamides P, Jodeleit H, Föhlinger M, Beigel F, Herbach N, Mueller T, Wolf E, Siebeck M, Gropp R. A mouse model for ulcerative colitis based on NOD-scid IL2R γnull mice reconstituted with peripheral blood mononuclear cells from affected individuals. Dis Model Mech 2016; 9:985-97. [PMID: 27491073 PMCID: PMC5047691 DOI: 10.1242/dmm.025452] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2016] [Accepted: 07/19/2016] [Indexed: 11/22/2022] Open
Abstract
Animal models reflective of ulcerative colitis (UC) remain a major challenge, and yet are crucial to understand mechanisms underlying the onset of disease and inflammatory characteristics of relapses and remission. Mouse models in which colitis-like symptoms are induced through challenge with toxins such as oxazolone, dextran sodium sulfate (DSS) or 2,4,6-trinitrobenzenesulfonic acid (TNBS) have been instrumental in understanding the inflammatory processes of UC. However, these neither reflect the heterogeneous symptoms observed in the UC-affected population nor can they be used to test the efficacy of inhibitors developed against human targets where high sequence and structural similarity of the respective ligands is lacking. In an attempt to overcome these problems, we have developed a mouse model that relies on NOD-scid IL2R γnull mice reconstituted with peripheral blood mononuclear cells derived from UC-affected individuals. Upon challenge with ethanol, mice developed colitis-like symptoms and changes in the colon architecture, characterized by influx of inflammatory cells, edema, crypt loss, crypt abscesses and epithelial hyperplasia, as previously observed in immune-competent mice. TARC, TGFβ1 and HGF expression increased in distal parts of the colon. Analysis of human leucocytes isolated from mouse spleen revealed an increase in frequencies of CD1a+, CD64+, CD163+ and TSLPR+ CD14+ monocytes, and antigen-experienced CD44+ CD4+ and CD8+ T-cells in response to ethanol. Analysis of human leucocytes from the colon of challenged mice identified CD14+ monocytes and CD11b+ monocytes as the predominant populations. Quantitative real-time PCR (RT-PCR) analysis from distal parts of the colon indicated that IFNγ might be one of the cytokines driving inflammation. Treatment with infliximab ameliorated symptoms and pathological manifestations, whereas pitrakinra had no therapeutic benefit. Thus, this model is partially reflective of the human disease and might help to increase the translation of animal and clinical studies. Summary: The phenotype and colitis-like symptoms induced in NSG mice reconstituted with PBMCs derived from ulcerative-colitis-affected donors reflect the human disease.
Collapse
Affiliation(s)
- Pia Palamides
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich 81377, Germany
| | - Henrika Jodeleit
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich 81377, Germany
| | - Michael Föhlinger
- Department of General- Visceral-, and Transplantation Surgery, Hospital of the University of Munich, Nussbaumstr. 20, Munich 80336, Germany
| | - Florian Beigel
- Department of Medicine II-Grosshadern, Ludwig Maximilians University, Munich, Germany
| | - Nadja Herbach
- Institute of Veterinary Pathology, Ludwig Maximilians University, Munich, Germany
| | - Thomas Mueller
- Julius von Sachs Institute, University of Würzburg, Würzburg 97082, Germany
| | - Eckhard Wolf
- Institute of Molecular Animal Breeding and Biotechnology, Gene Center, LMU Munich, Munich 81377, Germany Laboratory for Functional Genome Analysis (LAFUGA), Gene Center, LMU Munich, Munich 81377, Germany
| | - Matthias Siebeck
- Department of General- Visceral-, and Transplantation Surgery, Hospital of the University of Munich, Nussbaumstr. 20, Munich 80336, Germany
| | - Roswitha Gropp
- Department of General- Visceral-, and Transplantation Surgery, Hospital of the University of Munich, Nussbaumstr. 20, Munich 80336, Germany
| |
Collapse
|
109
|
Raad MA, Chams NH, Sharara AI. New and Evolving Immunotherapy in Inflammatory Bowel Disease. Inflamm Intest Dis 2016; 1:85-95. [PMID: 29922662 PMCID: PMC5988105 DOI: 10.1159/000445986] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/18/2016] [Accepted: 03/22/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Crohn's disease and ulcerative colitis are chronic inflammatory disorders associated with a dysregulated adaptive and innate immune response to gut commensals in genetically susceptible individuals. The pathogenesis of inflammatory bowel disease is complex, and the disease is characterized by significant phenotypic and genotypic heterogeneity. SUMMARY The introduction of anti-TNF biologics has resulted in improved clinical outcomes in patients with severe and moderately severe disease, but the current treatment paradigm continues to depend on systemic immunosuppression (steroids and immunomodulators) and surgical intervention in a significant number of patients, underscoring a significant unmet need. More recently, a number of genetic and immunologic abnormalities have been unraveled including aberrant intestinal mucosal defense function, abnormal intestinal permeability, dysregulated bacterial antigen processing by macrophages and presentation to T cells, cellular immune regulation and signaling, cytokine production, and leukocyte trafficking. KEY MESSAGES Understanding these molecular mechanisms and effector pathways presents an opportunity for the development of new and improved targeted therapies.
Collapse
Affiliation(s)
- Mohamad A. Raad
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | - Nour H. Chams
- School of Medicine, American University of Beirut, Beirut, Lebanon
| | - Ala I. Sharara
- Division of Gastroenterology, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| |
Collapse
|
110
|
Abstract
PURPOSE OF REVIEW Eosinophilic Esophagitis (EoE) is an emerging chronic atopic disease. Recent advances in understanding its genetic and molecular biology pathogenesis may lead to a better management of the disease RECENT FINDINGS EoE is an atopic disease. Most of the patients affected by EoE have other atopic diseases such as allergic rhinitis, asthma, IgE-mediated food allergies and/or atopic dermatitis. The local inflammation is a T helper type 2 (Th2) flogosis, which most likely is driven by a mixed IgE and n-IgE-mediated reaction to food and/or environmental allergens. Epidemiological studies show that EoE is an atopic disease with a strong genetic component. Genetic studies have shown that EoE is associated with single nucleotide polymorphism on genes, which are released by the epithelium and important in atopic inflammation such as thymic stromal lymphopoietin located (TSLP) close to the Th2 cytokine cluster [interleukin (IL)-4, IL-5, IL-13] on chromosome 5q22, Calpain 14, EMSY, and Eotaxin3. When the EoE diagnosis is made, it is imperative to control the local eosinophilic inflammation not only to give symptomatic relief to the patient, but also to prevent complications such as esophageal stricture and food impaction. SUMMARY EoE is treated like many other atopic diseases with a combination of topical steroids and/or food antigen avoidance. The new understanding of EoE may lead to more specific and definitive treatments of EoE.
Collapse
|
111
|
May RD, Fung M. Strategies targeting the IL-4/IL-13 axes in disease. Cytokine 2016; 75:89-116. [PMID: 26255210 DOI: 10.1016/j.cyto.2015.05.018] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2015] [Accepted: 05/15/2015] [Indexed: 02/07/2023]
Abstract
IL-4 and IL-13 are pleiotropic Th2 cytokines produced by a wide variety of different cell types and responsible for a broad range of biology and functions. Physiologically, Th2 cytokines are known to mediate host defense against parasites but they can also trigger disease if their activities are dysregulated. In this review we discuss the rationale for targeting the IL-4/IL-13 axes in asthma, atopic dermatitis, allergic rhinitis, COPD, cancer, inflammatory bowel disease, autoimmune disease and fibrotic disease as well as evaluating the associated clinical data derived from blocking IL-4, IL-13 or IL-4 and IL-13 together.
Collapse
|
112
|
Abstract
Current therapies for eosinophilic disorders are limited. Most treatment approaches remain empirical, are not supported by data from controlled clinical trials, involve the off-label use of agents developed for treatment of other diseases, and tend to rely heavily on the use of glucocorticoids and other agents with significant toxicity. Great progress has been made in the discovery, preclinical development, and clinical testing of a variety of biologics and small molecules that have the potential to directly or indirectly influence eosinophils, eosinophilic inflammation, and the consequences of eosinophil activation.
Collapse
|
113
|
Grainge CL, Maltby S, Gibson PG, Wark PAB, McDonald VM. Targeted therapeutics for severe refractory asthma: monoclonal antibodies. Expert Rev Clin Pharmacol 2016; 9:927-41. [DOI: 10.1586/17512433.2016.1172208] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Affiliation(s)
- Christopher L. Grainge
- Centre of Excellence in Severe Asthma, Hunter Medical Research Institute and Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Steven Maltby
- Centre of Excellence in Severe Asthma, Hunter Medical Research Institute and Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, Australia
| | - Peter G. Gibson
- Centre of Excellence in Severe Asthma, Hunter Medical Research Institute and Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Peter A. B. Wark
- Centre of Excellence in Severe Asthma, Hunter Medical Research Institute and Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| | - Vanessa M. McDonald
- Centre of Excellence in Severe Asthma, Hunter Medical Research Institute and Priority Research Centre for Healthy Lungs, The University of Newcastle, Newcastle, Australia
- Department of Respiratory and Sleep Medicine, John Hunter Hospital, Newcastle, Australia
| |
Collapse
|
114
|
Hisamatsu T, Erben U, Kühl AA. The Role of T-Cell Subsets in Chronic Inflammation in Celiac Disease and Inflammatory Bowel Disease Patients: More Common Mechanisms or More Differences? Inflamm Intest Dis 2016; 1:52-62. [PMID: 29922658 DOI: 10.1159/000445133] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2016] [Accepted: 03/02/2016] [Indexed: 12/13/2022] Open
Abstract
Background Chronic intestinal inflammation due to noninfectious causes represents a growing health issue all over the world. Celiac disease as well as inflammatory bowel diseases (IBD) like Crohn's disease and ulcerative and microscopic colitis involve uncontrolled T-cell activation and T-cell-mediated damage as common denominators. Therefore, diagnosis and treatment decisions clearly benefit from the knowledge of the intricacies of the systemic and the local T-cell activity. Summary Depending on the cytokine milieu, CD4+ T cells can differentiate into proinflammatory T helper 1 (Th1), anti-inflammatory Th2, antimicrobial Th17, pleiotropic Th9, tissue-instructing Th22 cells, and in the regulatory compartment forkhead box protein 3+ Treg, suppressive Tr1 or Th3 cells. Additionally, follicular Th cells provide B-cell help in antibody class switching; cytotoxic CD8+ T cells target virus-infected or tumor cells. This review discusses our current knowledge on the contribution of defined T-cell subpopulations to establishing and maintaining chronic intestinal inflammation in either of the above entities. It also puts emphasis on the differences in the prevalence of these diseases between Eastern and Western countries. Key Messages In celiac disease, the driving role of T cells in the lamina propria and in the epithelium mainly specific for two defined antigens is well established. Differences in genetics and lifestyle between Western and Eastern countries were instrumental in understanding underlying mechanisms. In IBD, the vast amount of potential antigens and the corresponding antigen-specific T cells makes it unlikely to find universal triggers. Increased mucosal CD4+ regulatory T cells in all four entities fail to control or abrogate local inflammatory processes. Thus, prevailing differences in the functional T-cell subtypes driving chronic intestinal inflammation in celiac disease and IBD at best allow some overlap in the treatment options for either disease.
Collapse
Affiliation(s)
- Tadakazu Hisamatsu
- The Third Department of Internal Medicine, Kyorin University School of Medicine, Mitaka, Tokyo, Japan
| | - Ulrike Erben
- Medical Department (Gastroenterology/Infectious Diseases/Rheumatology), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Research Center ImmunoSciences, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Anja A Kühl
- Medical Department (Gastroenterology/Infectious Diseases/Rheumatology), Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany.,Research Center ImmunoSciences, Campus Benjamin Franklin, Charité - Universitätsmedizin Berlin, Berlin, Germany
| |
Collapse
|
115
|
Tiwari A, Kasaian M, Heatherington AC, Jones HM, Hua F. A mechanistic PK/PD model for two anti-IL13 antibodies explains the difference in total IL-13 accumulation observed in clinical studies. MAbs 2016; 8:983-90. [PMID: 27049478 DOI: 10.1080/19420862.2016.1172151] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
IMA-638 and IMA-026 are humanized IgG1 monoclonal antibodies (mAbs) that target non-overlapping epitopes of IL-13. Separate first-in-human single ascending dose studies were conducted for each mAb. These studies had similar study designs, but mild to moderate asthmatics were recruited for the IMA-638 study and healthy subjects were recruited for the IMA-026 study. IMA-638 and IMA-026 showed similar pharmacokinetic (PK) profiles, but very different total IL-13 (free and drug bound IL-13) profiles; free IL13 was not measured. IMA-026 treatment induced a dose-dependent accumulation of total IL-13, while IMA-638 treatment led to a much smaller accumulation without any clear dose-response. To understand the differences between the two total IL-13 profiles and to predict the free IL-13 profiles for each mAb treatment, a mechanistic PK/pharmacodynamic model was developed. PK-related parameters were first fit to the mean PK profiles of each mAb separately; thereafter, the target-related parameters were fit to both total IL-13 profiles simultaneously. The IL-13 degradation rate was assumed to be the same for asthma patients and healthy subjects. The model suggests that an approximately 100× faster elimination of IL-13-IMA-638 complex than IL-13-IMA-026 complex could be responsible for the differences observed in total IL-13 profiles for the two mAbs. Furthermore, the model predicts that IMA-638 administration results in greater and more prolonged free IL-13 inhibition than equivalent dosing of IMA-026 despite similar binding KD and PK profile. In conclusion, joint modeling of two similar molecules provided mechanistic insight that the elimination rate of mAb-target complex can regulate the degree of free target inhibition.
Collapse
Affiliation(s)
- Abhinav Tiwari
- a Pharmacokinetics, Dynamics and Metabolism, New Biological Entities, Pfizer Inc. , Cambridge , MA , USA
| | - Marion Kasaian
- b Inflammation and Immunology Research Unit, Pfizer Inc. , Cambridge , MA , USA
| | - Anne C Heatherington
- c Quantitative Clinical Sciences, PharmaTherapeutcis R&D, Pfizer Inc. , Cambridge , MA , USA
| | - Hannah M Jones
- a Pharmacokinetics, Dynamics and Metabolism, New Biological Entities, Pfizer Inc. , Cambridge , MA , USA
| | - Fei Hua
- c Quantitative Clinical Sciences, PharmaTherapeutcis R&D, Pfizer Inc. , Cambridge , MA , USA
| |
Collapse
|
116
|
De Salvo C, Wang XM, Pastorelli L, Mattioli B, Omenetti S, Buela KA, Chowdhry S, Garg RR, Goodman WA, Rodriguez-Palacios A, Smith DE, Abbott DW, Cominelli F, Bamias G, Xin W, Lee JJ, Vecchi M, Pizarro TT. IL-33 Drives Eosinophil Infiltration and Pathogenic Type 2 Helper T-Cell Immune Responses Leading to Chronic Experimental Ileitis. THE AMERICAN JOURNAL OF PATHOLOGY 2016; 186:885-898. [PMID: 26908008 PMCID: PMC5807926 DOI: 10.1016/j.ajpath.2015.11.028] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Revised: 11/03/2015] [Accepted: 11/19/2015] [Indexed: 02/08/2023]
Abstract
Although a clear association has been established between IL-33 and inflammatory bowel disease, mechanistic studies to date, primarily using acute murine models of colitis, have yielded contradicting results, demonstrating both pathogenic and protective roles. We used a well-characterized, spontaneous model of inflammatory bowel disease [ie, SAMP1/YitFc (SAMP) mice] to investigate the role of IL-33 during chronic intestinal inflammation. Our results showed marked eosinophil infiltration into the gut mucosa with increased levels of eotaxins and type 2 helper T-cell (Th2) cytokines as disease progressed and became more severe, which could be reversed upon either eosinophil depletion or blockade of IL-33 signaling. Exogenous IL-33 administration recapitulated these effects in ilea of uninflamed (parental) control AKR/J mice. Human data supported these findings, showing colocalization and up-regulation of IL-33 and eosinophils in the colonic mucosa of inflammatory bowel disease patients versus noninflamed controls. Finally, colonization of commensal flora by fecal material transplantation into germ-free SAMP and the presence of the gut microbiome induced IL-33, subsequent eosinophil infiltration, and mounting of Th2 immune responses, leading to exacerbation of chronic intestinal inflammation characteristic of SAMP mice. These data demonstrate a pathogenic role for IL-33-mediated eosinophilia and activation of Th2 immunity in chronic intestinal inflammation that is dependent on the gut microbiome. Targeting IL-33 may represent a novel therapeutic approach to treat patients with inflammatory bowel disease.
Collapse
Affiliation(s)
- Carlo De Salvo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Xiao-Ming Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Luca Pastorelli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Benedetta Mattioli
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Sara Omenetti
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Kristine A Buela
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Saleem Chowdhry
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Rekha R Garg
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Wendy A Goodman
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | | | - Dirk E Smith
- Inflammation Research, Amgen, Seattle, Washington
| | - Derek W Abbott
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - Giorgos Bamias
- Academic Department of Gastroenterology, Kapodistrian University of Athens and Laikon Hospital, Athens, Greece
| | - Wei Xin
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio
| | - James J Lee
- Division of Pulmonary Medicine, Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Scottsdale, Arizona
| | - Maurizio Vecchi
- Gastroenterology and Gastrointestinal Endoscopy Unit, IRCCS Policlinico San Donato, San Donato Milanese and Department of Biomedical Sciences, University of Milan, Milan, Italy
| | - Theresa T Pizarro
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio.
| |
Collapse
|
117
|
The Link between the Appendix and Ulcerative Colitis: Clinical Relevance and Potential Immunological Mechanisms. Am J Gastroenterol 2016; 111:163-9. [PMID: 26416189 DOI: 10.1038/ajg.2015.301] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 08/11/2015] [Indexed: 12/11/2022]
Abstract
The human appendix has long been considered as a vestigial organ, an organ that has lost its function during evolution. In recent years, however, reports have emerged that link the appendix to numerous immunological functions in humans. Evidence has been presented for an important role of the appendix in maintaining intestinal health. This theory suggests that the appendix may be a reservoir or 'safe house' from which the commensal gut flora can rapidly be reestablished if it is eradicated from the colon. However, the appendix may also have a role in the development of inflammatory bowel disease (IBD). Several large epidemiological cohort studies have demonstrated the preventive effect of appendectomy on the development of ulcerative colitis, a finding that has been confirmed in murine colitis models. In addition, current studies are examining the possible therapeutic effect of an appendectomy to modulate disease course in patients with ulcerative colitis. This literature review assesses the current knowledge about the clinical and immunological aspects of the vermiform appendix in IBD and suggests that the idea of the appendix as a vestigial remnant should be discarded.
Collapse
|
118
|
Bamias G, Pizarro TT, Cominelli F. Pathway-based approaches to the treatment of inflammatory bowel disease. Transl Res 2016; 167:104-115. [PMID: 26408803 PMCID: PMC4782917 DOI: 10.1016/j.trsl.2015.09.002] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Revised: 09/01/2015] [Accepted: 09/02/2015] [Indexed: 12/17/2022]
Abstract
Crohn's disease and ulcerative colitis, collectively termed inflammatory bowel disease (IBD), are immunologic disorders that represent the prototypes of chronic intestinal inflammation. Their pathogenesis involves the dysregulated interaction between the intestinal microbiota and the gut-associated mucosal immune system that takes place when genetically predisposed individuals are exposed to detrimental environmental triggers. In recent years, the therapeutic dogma in IBD has shifted away from the administration of nonspecific immunosuppressives toward a pathway-based approach. In this review, we present an outlook of IBD treatment based on this new conceptual approach. Firstly, we will provide an overview of the major aspects of IBD pathogenesis with emphasis on specific pathway-based defects. Secondly, we will examine in detail the development of novel therapeutic approaches that can be used to target genetics, dysbiosis, the epithelial barrier, proinflammatory cytokines, and leukocyte trafficking. Most of these strategies are still in the developmental phase, but promising approaches include fecal microbiota transplantation as a means to correct IBD-related dysbiosis; administration of modified phosphatidylcholine to enhance the function of the intestinal mucous and tighten the defective epithelial barrier; the reduction of over-reactive proinflammatory pathways through the blockade of novel, nontumor necrosis factor inflammatory mediators via monoclonal antibodies against the common p40 chain of interleukin (IL-12) and IL-23, Janus kinase inhibitors, or antisense oligonucleotides against inhibitors of the immunosuppressive cytokine transforming growth factor-β1; and finally, inhibition of leukocyte trafficking to the gut via neutralization of the gut-specific α4β7 integrin. Availability of such diverse treatment modalities with specific pathway-based targets will increase the therapeutic options for patients with IBD.
Collapse
Affiliation(s)
- Giorgos Bamias
- First Department of Gastroenterology, Ethnikon and Kapodistriakon University of Athens, Laikon Hospital, Athens, Greece
| | - Theresa T Pizarro
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio
| | - Fabio Cominelli
- Department of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio; Digestive Health Research Institute, Case Western Reserve University, Cleveland, Ohio.
| |
Collapse
|
119
|
Atreya R, Billmeier U, Rath T, Mudter J, Vieth M, Neumann H, Neurath MF. First case report of exacerbated ulcerative colitis after anti-interleukin-6R salvage therapy. World J Gastroenterol 2015; 21:12963-12969. [PMID: 26668517 PMCID: PMC4671048 DOI: 10.3748/wjg.v21.i45.12963] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Revised: 06/30/2015] [Accepted: 09/14/2015] [Indexed: 02/06/2023] Open
Abstract
We present the case of a 53-year-old woman with long-standing ulcerative colitis and severe, steroid-dependent disease course unresponsive to treatment with azathioprine, methotrexate, anti-TNF antibodies (infliximab, adalimumab) and tacrolimus, who refused colectomy as a therapeutic option. As the pro-inflammatory cytokine interleukin-6 (IL-6) had been identified as a crucial regulator in the immunopathogenesis of inflammatory bowel diseases, we treated the patient with biweekly intravenous infusions of an anti-IL-6R antibody (tocilizumab) for 12 wk. However, no clinical improvement of disease activity was noted. In fact, endoscopic, histological and endomicroscopic assessment demonstrated exacerbation of mucosal inflammation and ulcer formation upon anti-IL-6R therapy. Mechanistic studies revealed that tocilizumab treatment failed to suppress intestinal IL-6 production, impaired epithelial barrier function and induced production of pro-inflammatory cytokines such as TNF, IL-21 and IFN-γ. Inhibition of IL-6 by tocilizumab had no clinical benefit in this patient with intractable ulcerative colitis and even led to exacerbation of mucosal inflammation. Our findings suggest that anti-IL-6R antibody therapy may lead to aggravation of anti-TNF resistant ulcerative colitis. When targeting IL-6, the differential responsiveness of target cells has to be taken into account, as IL-6 on the one side promotes acute and chronic mucosal inflammation via soluble IL-6R signaling but on the other side also strongly contributes to epithelial cell survival via membrane bound IL-6R signaling.
Collapse
MESH Headings
- Anti-Inflammatory Agents/administration & dosage
- Anti-Inflammatory Agents/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/adverse effects
- Biopsy
- Colitis, Ulcerative/diagnosis
- Colitis, Ulcerative/drug therapy
- Colitis, Ulcerative/genetics
- Colitis, Ulcerative/immunology
- Colonoscopy
- Cytokines/genetics
- Cytokines/immunology
- Disease Progression
- Female
- Gastrointestinal Agents/administration & dosage
- Gastrointestinal Agents/adverse effects
- Humans
- Middle Aged
- Molecular Targeted Therapy
- Receptors, Interleukin-6/antagonists & inhibitors
- Receptors, Interleukin-6/immunology
- Treatment Outcome
Collapse
|
120
|
New insights into immune mechanisms underlying autoimmune diseases of the gastrointestinal tract. Autoimmun Rev 2015; 14:1161-9. [PMID: 26275585 DOI: 10.1016/j.autrev.2015.08.004] [Citation(s) in RCA: 90] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2015] [Accepted: 08/05/2015] [Indexed: 02/07/2023]
|
121
|
Crispín JC, Rosetti F, Hernández-Molina G. Lessons from Sjögren’s syndrome etiopathogenesis: Novel cellular and molecular targets. World J Immunol 2015; 5:152-159. [DOI: 10.5411/wji.v5.i3.152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2015] [Revised: 09/22/2015] [Accepted: 11/17/2015] [Indexed: 02/05/2023] Open
Abstract
Sjögren’s syndrome (SS) is a systemic autoimmune disease that affects primarily the lacrimal and salivary glands. In addition to a systemic autoimmune response directed against ubiquitous antigens (such as Ro and La antigens), patients with SS mount a localized response that affects the epithelial component of exocrine glands leading to the establishment of a destructive inflammatory infiltrate comprised of activated T and B cells. Local chemokine and cytokine production drive the recruitment and local activation of immune cells that cause injury to acinar cells. CD4 T cells with different functional differentiation programs including Th1 (IFN-γ), Th2 (IL-13, IL-4) and Th17 (IL-17, IL-21, IL-22) as well as diverse cytokine signaling pathways, are involved at the initiation, perpetuation, and progression of the disease. Which factors initiate this response and allow it to become chronic are unknown. Proposed mechanisms include viral infections and acinar cell apoptosis. Moreover risk-conferring genetic variants, probably through the facilitation of innate and adaptive immune activation, most certainly contribute to the creation of an underlying environment that fosters tolerance loss and facilitates perpetuation of the autoimmune response. In this review, we describe the mechanisms through which the immune response causes SS and emphasize the pathways that are amenable of being targeted with therapeutic purposes.
Collapse
|
122
|
Abstract
PURPOSE OF REVIEW Type 2 (Th2) immune responses play important roles in intestinal immunity by contributing to the maintenance of mucosal homeostasis, not only conferring protection against helminthic infection but also participating in pro-inflammatory pathways in chronic intestinal inflammatory disorders, including inflammatory bowel disease. The current review focuses on recent developments regarding the role of Th2 responses in intestinal inflammation. RECENT FINDINGS Th2 gut mucosal responses are promoted by mediators that are released following injury to the epithelium, and act as alarmin-type danger signals. Interleukin (IL)-33 is prominent among such factors and demonstrates a dichotomous function, exerting either protective or pro-inflammatory effects, depending on its cellular compartmentalization. The pool of type 2 effector cells has been enriched recently to include not only classical CD4+ Th2 lymphocytes but also a subset of innate lymphocytes (ILC2s) that express the transcriptional factor GATA binding protein 3 and secrete IL-4, IL-5, and IL-13. ILC2s play important roles during infection with helminths and bi-directionally interact with Th2 CD4+ lymphocytes, thus establishing a transition from innate to adaptive immunological pathways. Th2 responses are also involved in pro-inflammatory pathways at the intestinal mucosa, and neutralization of the pivotal cytokines IL-4 and IL-13 has been shown to regulate experimental intestinal inflammation. In striking contrast, however, neutralization of human IL-13 had no therapeutic effect in patients with ulcerative colitis. SUMMARY Further studies will be required to delineate the specific mechanisms of type 2 mucosal immunity in inflammatory bowel disease and examine the applicability of Th2-targeted therapies for intestinal inflammation.
Collapse
Affiliation(s)
- Giorgos Bamias
- Academic Department of Gastroenterology, Kapodistrian University of Athens, Laikon Hospital, Athens, 11527, Greece
| | - Fabio Cominelli
- Division of Gastrointestinal and Liver Disease, Case Western Reserve University, School of Medicine, Cleveland, OH, 44106, USA
| |
Collapse
|
123
|
Gómez-Gómez GJ, Masedo &A, Yela C, Martínez-Montiel MDP, Casís B. Current stage in inflammatory bowel disease: What is next? World J Gastroenterol 2015; 21:11282-11303. [PMID: 26525013 PMCID: PMC4616205 DOI: 10.3748/wjg.v21.i40.11282] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 07/12/2015] [Accepted: 09/02/2015] [Indexed: 02/06/2023] Open
Abstract
In recent years, the incidence of inflammatory bowel disease (IBD) has been on the rise, extending to countries where it was infrequent in the past. As a result, the gap between high and low incidence countries is decreasing. The disease, therefore, has an important economic impact on the healthcare system. Advances in recent years in pharmacogenetics and clinical pharmacology have allowed for the development of treatment strategies adjusted to the patient profile. Concurrently, new drugs aimed at inflammatory targets have been developed that may expand future treatment options. This review examines advances in the optimization of existing drug treatments and the development of novel treatment options for IBD.
Collapse
|
124
|
Abstract
After a relatively long time of failed developments and negative clinical trials in pharmacological inflammatory bowel disease (IBD) therapy we now phase a time of a great number of successful studies and new therapy principles that will most likely make it into clinical practice. This will change the landscape of IBD therapy in future markedly. Many new therapeutic principles have been developed and old ones that seemed to have failed such as anti-sense technology suddenly now provide promising results. Some initially promising therapies will need further development or have failed such as Trichuris suis ova therapy (but not helminth therapy in general), CCR9 targeted therapies or recombinant IL-10. In contrast anti-leukocate trafficking therapies appear to be quite promising. Vedolizumab is the first in class anti-integrin antibody that was approved for the therapy of CD and UC recently. Other anti-integrin antibodies and small molecule adhesion inhibitors will most likely be approved in the next years for IBD therapy. Tofacitinib, a small molecule JAK inhibitor, is a promising candidate for the treatment of UC. Phosphatidylcholine may be a future option for patients with 5-ASA refractory UC or 5-ASA intolerance. The preliminary data for Mongersen, a Smad7 antisense oligonucleotide, are promising despite some concerns about long term effect of TGFβ induction. Anti IL6 strategies will hopefully be further evaluated keeping in mind the caveat of a lack of CRP induction in anti-IL6 treated patients. Stem cell transplantation will become an option for patients that have experienced failure of established medications. Fecal microbiota transplantation and also perhaps combined probiotic therapy is a field that will be evaluated in more detail in the near future especially for UC patients. Based on these new developments treatment algorithms need to be updated. This review will reflect these current developments and give a perspective for future IBD therapy.
Collapse
|
125
|
Maselli DJ, Keyt H, Rogers L. Profile of lebrikizumab and its potential in the treatment of asthma. J Asthma Allergy 2015; 8:87-92. [PMID: 26309415 PMCID: PMC4539078 DOI: 10.2147/jaa.s69932] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Interleukin (IL)-13 has been associated with multiple inflammatory features of asthma. It affects multiple cellular lines in asthma and is a key mediator in airway hyperreactivity and remodeling. Periostin, an extracellular protein, has been used as a surrogate marker of IL-13 activity and has been linked to airway remodeling by inducing subepithelial fibrosis. Lebrikizumab is a humanized monoclonal antibody that targets IL-13. Studies have demonstrated promising results with lebrikizumab therapy in asthma with regard to pulmonary function and exacerbation rates, especially on those patients with surrogate markers of T helper cell type 2-driven inflammation (ie, elevated immunoglobulin E levels, eosinophil counts, periostin levels). Lebrikizumab appears to be a safe therapy, but there are ongoing studies evaluating its efficacy and safety profile. Other therapies that target IL-13 and the receptor of IL-4/IL-13 have been studied, but future studies are needed to determine their role in the treatment of asthma.
Collapse
Affiliation(s)
- Diego Jose Maselli
- Department of Medicine, Division of Pulmonary Diseases and Critical Care, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Holly Keyt
- Department of Medicine, Division of Pulmonary Diseases and Critical Care, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Linda Rogers
- Pulmonary, Critical Care, and Sleep Medicine Division, Mount Sinai-National Jewish Health Respiratory Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
126
|
Bürger M, Schmidt C, Teich N, Stallmach A. Medical Therapy of Active Ulcerative Colitis. VISZERALMEDIZIN 2015; 31:236-45. [PMID: 26557831 PMCID: PMC4608602 DOI: 10.1159/000436959] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Background Medical therapy of mild and moderate ulcerative colitis (UC) of any extent is evidence-based and standardized by national and international guidelines. However, patients with steroid-refractory UC still represent a challenge. Methods A literature search using PubMed (search terms: ulcerative colitis, therapy, new, 1-2008-2015) resulted in 821 publications. For the current article, 88 citations were extracted including 36 randomized controlled studies, 18 reviews, and 8 meta-analyses. Results In steroid-refractory UC, early intensive therapy using anti-tumor necrosis factor (TNF) antibodies or the calcineurin inhibitors cyclosporine and tacrolimus is indicated in any case to prevent progression to a toxic megacolon and/or to avoid proctocolectomy. In patients with chronic disease activity, treatment with anti-TNF antibodies has a higher level of evidence than azathioprine therapy and should therefore be preferred. However, there is a subgroup of UC patients who may achieve prolonged steroid-free remission on azathioprine monotherapy. The importance of vedolizumab, a newly registered inhibiting antibody against integrin, has not yet been fully clarified since direct comparison studies are lacking, in particular in relation to anti-TNF antibodies. Conclusion There is a great need for additional innovative therapies, especially in cases of primary non-response or secondary loss of response to anti-TNF antibodies. New small molecules (Janus kinase inhibitors) are promising with an acceptable safety profile and efficacy in UC. Further, strategies that target the intestinal microbiome are currently considered for patients with active or relapsing UC, and may in the future open up new therapeutic options.
Collapse
Affiliation(s)
- Martin Bürger
- Department of Internal Medicine IV, Jena University Hospital, Jena, Germany
| | - Carsten Schmidt
- Department of Internal Medicine IV, Jena University Hospital, Jena, Germany
| | - Niels Teich
- Group Practice for Digestive and Metabolic Diseases, Leipzig, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV, Jena University Hospital, Jena, Germany
| |
Collapse
|
127
|
Hua F, Ribbing J, Reinisch W, Cataldi F, Martin S. A pharmacokinetic comparison of anrukinzumab, an anti- IL-13 monoclonal antibody, among healthy volunteers, asthma and ulcerative colitis patients. Br J Clin Pharmacol 2015; 80:101-9. [PMID: 25614144 PMCID: PMC4500329 DOI: 10.1111/bcp.12589] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2014] [Revised: 12/18/2014] [Accepted: 01/03/2015] [Indexed: 01/08/2023] Open
Abstract
AIMS Anrukinzumab is an anti-IL13 monoclonal antibody. The goals of this study are to characterize the pharmacokinetics of anrukinzumab in healthy volunteers and different disease states and to identify covariates. METHODS A population pharmacokinetic (PK) model was developed in NONMEM, using data from five clinical studies including healthy volunteers, asthma and ulcerative colitis (UC) patients. Different dosing regimens including different routes of administration were also included in the data. RESULTS The PK of anrukinzumab were described by a two compartment model with first order absorption and elimination. The population estimates (relative standard error) of the volumes of distribution in the central (Vc ) and peripheral (Vp ) compartments were 3.8 (4.6%) and 2.2 l (8.7%), respectively. In non-UC patients, the population estimate of the systemic clearance (CL) and inter-compartmental CL was 0.00732 l h(-1) (4.9%) and 0.0224 l h(-1) (15.4%). For subcutaneous administration, the absorption rate constant was 0.012 h(-1) (6.6%) and bioavailability was nearly 100% in healthy and mild to moderate asthma patients. Both V and CL increased with body weight. CL (but not V) decreased with increasing baseline albumin concentrations. UC patients had an increased CL of 72.3% (10.5%), after correction for differences in body weight and albumin. Moderate to severe asthma patients had decreased bioavailability compared with other populations. CONCLUSIONS Anrukinzumab's PK behave like a typical antibody. UC patients were identified to have a faster CL of anrukinzumab than healthy volunteers and asthma patients. This finding suggests a higher dose level may be required for this population.
Collapse
Affiliation(s)
- Fei Hua
- BioTx Clinical Research, Pfizer Inc.Cambridge, MA, USA
| | | | | | - Fabio Cataldi
- BioTx Clinical Research, Pfizer Inc.Cambridge, MA, USA
| | - Steven Martin
- Global Pharmacometrics, Pfizer Inc.Cambridge, MA, USA
| |
Collapse
|