1
|
Kajikawa G, Sawada T, Nakamura M, Yamamura T, Maeda K, Ishikawa E, Uetsuki K, Hirose T, Iida T, Mizutani Y, Yamao K, Ishikawa T, Furukawa K, Kawashima H. Predictors of the efficacy of vedolizumab in patients with ulcerative colitis. NAGOYA JOURNAL OF MEDICAL SCIENCE 2024; 86:407-421. [PMID: 39355361 PMCID: PMC11439607 DOI: 10.18999/nagjms.86.3.407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 12/08/2023] [Indexed: 10/03/2024]
Abstract
Vedolizumab is a treatment option for ulcerative colitis but data on predictors of treatment response remain insufficient to establish personalized treatment strategies. We aimed to investigate the real-world effectiveness of vedolizumab in adult patients with ulcerative colitis and explore factors involved in predicting treatment response. This single-center, single-arm, prospective observational study included 26 patients with clinically active ulcerative colitis patients' characteristics at baseline, epidemiological information, existing treatment, clinical activity index score, endoscopic score, and blood test data were collected. Serum levels of tumor necrosis factors alpha, interferon gamma, interleukin-4, interleukin-6, interleukin-10, interleukin-17, soluble mucosal addressin cell adhesion molecule 1, and soluble vascular cell adhesion molecule 1 were measured. Patient characteristics in the remission and non-remission groups were compared based on these parameters. Clinical remission at 6 weeks of treatment occurred in 9 (35%) of the 26 patients. At 14 weeks, clinical remission was observed in 11 patients (42%). There were no significant differences pertaining to age, sex, duration of disease, extent of disease, steroid resistance, or prior treatment with biological agents among the two groups after 14 weeks of treatment. Hemoglobin ≥ 11.5 g/dL (odds ratio, 15.0; 95% confidence interval, 1.50-149; P=0.014) and soluble mucosal addressin cell adhesion molecule 1 ≥ 765 pg/mL (odds ratio, 17.3; 95% confidence interval, 2.36-127; P=0.004) were significant factors. In conclusion, hemoglobin and serum soluble mucosal addressin cell adhesion molecule 1 levels are factors correlated with the therapeutic efficacy of vedolizumab.
Collapse
Affiliation(s)
- Go Kajikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Tsunaki Sawada
- Department of Endoscopy, Nagoya University Hospital, Nagoya, Japan
| | - Masanao Nakamura
- Department of Endoscopy, Nagoya University Hospital, Nagoya, Japan
| | - Takeshi Yamamura
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Keiko Maeda
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Eri Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kota Uetsuki
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Hirose
- Department of Endoscopy, Nagoya University Hospital, Nagoya, Japan
| | - Tadashi Iida
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yasuyuki Mizutani
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kentaro Yamao
- Department of Endoscopy, Nagoya University Hospital, Nagoya, Japan
| | - Takuya Ishikawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Kazuhiro Furukawa
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
2
|
Palandurkar GS, Kumar S. Biofilm's Impact on Inflammatory Bowel Diseases. Cureus 2023; 15:e45510. [PMID: 37868553 PMCID: PMC10585119 DOI: 10.7759/cureus.45510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2023] [Accepted: 09/18/2023] [Indexed: 10/24/2023] Open
Abstract
The colon has a large surface area covered with a thick mucus coating. Colon's biomass consists of about 1,012 colony-forming units per gram of feces and 500-1,000 distinct bacterial species. The term inflammatory bowel disease (IBD) indicates the collection of intestinal illnesses in which the digestive system (esophagus, large intestine, mouth, stomach, and small intestine) experiences persistent inflammation. IBD development is influenced by environmental (infections, stress, and nutrition) and genetic factors. The microbes present in gut microbiota help maintain intestinal homeostasis and support immune and epithelial cell growth, differentiation, as well as proliferation. It has been discovered that a variety of variables and microorganisms are crucial for the development of biofilms and mucosal colonization during IBD. An extracellular matrix formed by bacteria supports biofilm production in our digestive system and harms the host's immunological response. Irritable bowel syndrome (IBS) and IBD considerably affect human socioeconomic well-being and the standard of living. IBD is a serious public health issue, affecting millions of people across the globe. The gut microbiome may significantly influence IBS pathogenesis, even though few diagnostic and treatment options are available. As a result, current research focuses more on disrupting biofilm in IBD patients and stresses primarily on drugs that help improve the quality of life for human well-being. We evaluate studies on IBD and bacterial biofilm to add fresh insights into the existing state of knowledge of biofilm formation in IBD, incidence of IBD patients, molecular level of investigations, bacteria that are involved in the formation of biofilm, and present and down the line regimens and probiotics. Planning advanced ways to control and eradicate bacteria in biofilms should be the primary goal to add fresh insights into generating innovative diagnostic and alternative therapy options for IBD.
Collapse
Affiliation(s)
- Gopal S Palandurkar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| | - Sunil Kumar
- Department of Medicine, Jawaharlal Nehru Medical College, Datta Meghe Institute of Higher Education and Research, Wardha, IND
| |
Collapse
|
3
|
Feliu V, Gomez-Roca C, Michelas M, Thébault N, Lauzéral-Vizcaino F, Salvioni A, Scandella L, Sarot E, Valle C, Balança CC, Scarlata CM, Delord JP, Ayyoub M, Devaud C. Distant antimetastatic effect of enterotropic colon cancer-derived α4β7 +CD8 + T cells. Sci Immunol 2023; 8:eadg8841. [PMID: 37289857 DOI: 10.1126/sciimmunol.adg8841] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2023] [Accepted: 03/29/2023] [Indexed: 06/10/2023]
Abstract
Despite the high prognostic value of immune infiltrates in colorectal cancer (CRC), metastatic disease remains resistant to immunotherapy by immune checkpoint blockade (ICB). Here, we show, in metastatic CRC preclinical models, that orthotopically implanted primary colon tumors exert a colon-specific antimetastatic effect on distant hepatic lesions. Enterotropic α4β7 integrin-expressing neoantigen-specific CD8 T cells were key components of the antimetastatic effect. Accordingly, the presence of concomitant colon tumors improved control of liver lesions by anti-PD-L1 proof-of-concept immunotherapy and generated protective immune memory, whereas partial depletion of α4β7+ cells abrogated control of metastases. Last, in patients with metastatic CRC, response to ICB was associated with expression of α4β7 integrin in metastases and with circulating α4β7+ CD8 T cells. Our findings identify a systemic cancer immunosurveillance role for gut-primed tumor-specific α4β7+ CD8 T cells.
Collapse
Affiliation(s)
- Virginie Feliu
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Carlos Gomez-Roca
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Marie Michelas
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Noémie Thébault
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Françoise Lauzéral-Vizcaino
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Anna Salvioni
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Lise Scandella
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Emeline Sarot
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Carine Valle
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
| | - Camille-Charlotte Balança
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Clara-Maria Scarlata
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Jean-Pierre Delord
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Maha Ayyoub
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| | - Christel Devaud
- Université de Toulouse, Inserm, CNRS, Université Toulouse III-Paul Sabatier, Centre de Recherches en Cancérologie de Toulouse, Toulouse, France
- Institut Universitaire du Cancer de Toulouse (IUCT)-Oncopole, Institut Claudius Regaud, Toulouse, France
| |
Collapse
|
4
|
Tackling Inflammatory Bowel Diseases: Targeting Proinflammatory Cytokines and Lymphocyte Homing. Pharmaceuticals (Basel) 2022; 15:ph15091080. [PMID: 36145301 PMCID: PMC9502105 DOI: 10.3390/ph15091080] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/23/2022] [Accepted: 08/26/2022] [Indexed: 11/21/2022] Open
Abstract
Inflammatory bowel diseases (IBDs) are characterized by chronic inflammatory disorders that are a result of an abnormal immune response mediated by a cytokine storm and immune cell infiltration. Proinflammatory cytokine therapeutic agents, represented by TNF inhibitors, have developed rapidly over recent years and are promising options for treating IBD. Antagonizing interleukins, interferons, and Janus kinases have demonstrated their respective advantages in clinical trials and are candidates for anti-TNF therapeutic failure. Furthermore, the blockade of lymphocyte homing contributes to the excessive immune response in colitis and ameliorates inflammation and tissue damage. Factors such as integrins, selectins, and chemokines jointly coordinate the accumulation of immune cells in inflammatory regions. This review assembles the major targets and agents currently targeting proinflammatory cytokines and lymphatic trafficking to facilitate subsequent drug development.
Collapse
|
5
|
Wyatt NJ, Speight RA, Stewart CJ, Kirby JA, Lamb CA. Targeting Leukocyte Trafficking in Inflammatory Bowel Disease. BioDrugs 2021; 35:473-503. [PMID: 34613592 DOI: 10.1007/s40259-021-00496-5] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/04/2021] [Indexed: 12/11/2022]
Abstract
In the last two decades, understanding of inflammatory bowel disease (IBD) immunopathogenesis has expanded considerably. Histopathological examination of the intestinal mucosa in IBD demonstrates the presence of a chronic inflammatory cell infiltrate. Research has focused on identifying mechanisms of immune cell trafficking to the gastrointestinal tract that may represent effective gut-selective targets for IBD therapy whilst avoiding systemic immunosuppression that may be associated with off-target adverse effects such as infection and malignancy. Integrins are cell surface receptors that can bind to cellular adhesion molecules to mediate both leukocyte homing and retention. In 2014, Vedolizumab (Entyvio®) was the first anti-integrin (anti-α4ß7 monoclonal antibody) treatment to be approved for use in IBD. Several other anti-integrin therapies are currently in advanced stages of development, including novel orally administered small-molecule drugs. Drugs targeting alternative trafficking mechanisms such as mucosal addressin cellular adhesion molecule-1 and sphingosine-1-phosphate receptors are also being evaluated. Here, we summarise key established and emerging therapies targeting leukocyte trafficking that may play an important role in realising the goal of stratified precision medicine in IBD care.
Collapse
Affiliation(s)
- Nicola J Wyatt
- Faculty of Medical Sciences, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK
| | - R Alexander Speight
- Faculty of Medical Sciences, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.,Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK
| | - Christopher J Stewart
- Faculty of Medical Sciences, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - John A Kirby
- Faculty of Medical Sciences, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK
| | - Christopher A Lamb
- Faculty of Medical Sciences, Translational & Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK. .,Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, NE1 4LP, UK.
| |
Collapse
|
6
|
Garlatti V, Lovisa S, Danese S, Vetrano S. The Multiple Faces of Integrin-ECM Interactions in Inflammatory Bowel Disease. Int J Mol Sci 2021; 22:10439. [PMID: 34638778 PMCID: PMC8508809 DOI: 10.3390/ijms221910439] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 12/03/2022] Open
Abstract
Inflammatory Bowel Disease (IBD) comprises a series of chronic and relapsing intestinal diseases, with Crohn's disease and ulcerative colitis being the most common. The abundant and uncontrolled deposition of extracellular matrix, namely fibrosis, is one of the major hallmarks of IBD and is responsible for the progressive narrowing and closure of the intestine, defined as stenosis. Although fibrosis is usually considered the product of chronic inflammation, the substantial failure of anti-inflammatory therapies to target and reduce fibrosis in IBD suggests that fibrosis might be sustained in an inflammation-independent manner. Pharmacological therapies targeting integrins have recently shown great promise in the treatment of IBD. The efficacy of these therapies mainly relies on their capacity to target the integrin-mediated recruitment and functionality of the immune cells at the damage site. However, by nature, integrins also act as mechanosensitive molecules involved in the intracellular transduction of signals and modifications originating from the extracellular matrix. Therefore, understanding integrin signaling in the context of IBD may offer important insights into mechanisms of matrix remodeling, which are uncoupled from inflammation and could underlie the onset and persistency of intestinal fibrosis. In this review, we present the currently available knowledge on the role of integrins in the etiopathogenesis of IBD, highlighting their role in the context of immune-dependent and independent mechanisms.
Collapse
Affiliation(s)
- Valentina Garlatti
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Pharmaceutical Sciences, University of Piemonte Orientale ‘A. Avogadro’, 28100 Novara, Italy
| | - Sara Lovisa
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Silvio Danese
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| | - Stefania Vetrano
- IRCCS Humanitas Research Hospital, Rozzano, 20089 Milan, Italy; (V.G.); (S.L.); (S.D.)
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, 20072 Milan, Italy
| |
Collapse
|
7
|
Gubatan J, Keyashian K, Rubin SJS, Wang J, Buckman CA, Sinha S. Anti-Integrins for the Treatment of Inflammatory Bowel Disease: Current Evidence and Perspectives. Clin Exp Gastroenterol 2021; 14:333-342. [PMID: 34466013 PMCID: PMC8402953 DOI: 10.2147/ceg.s293272] [Citation(s) in RCA: 50] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 08/10/2021] [Indexed: 12/15/2022] Open
Abstract
Leukocyte trafficking to the gastrointestinal tract is recognized to play a role in the pathogenesis of inflammatory bowel disease (IBD). Integrins are expressed on immune cells and interact with cell adhesion molecules (CAM) to mediate leukocyte trafficking. Blockade of the gut-tropic integrin α4β7 and its subunits has been exploited as a therapeutic target in IBD. Natalizumab (anti-α4) is approved for moderate to severe Crohn's disease (CD), but its use is limited due to potential risk of progressive multifocal leukoencephalopathy. Vedolizumab (anti-α4β7) is approved for the treatment of ulcerative colitis (UC) and CD. It is the most widely used anti-integrin therapy in IBD and has been shown to be effective in both induction and maintenance therapy, with a favorable safety profile. Several models incorporating clinical, genetic, immune, gut microbial, and vitamin D markers to predict response to vedolizumab in IBD have been developed. Etrolizumab (anti-β7) blocks leukocyte trafficking via α4β7 and cell adhesion via αEβ7 integrins. Large phase 3 clinical trials evaluating efficacy of etrolizumab in the induction and maintenance of patients with IBD are underway. Other investigational anti-integrin therapies include abrilumab (anti-α4β7 IgG2), PN-943 (orally administered and gut-restricted α4β7 antagonist peptide), AJM300 (orally active small molecule inhibitor of α4), and ontamalimab (anti-MAdCAM-1 IgG).
Collapse
Affiliation(s)
- John Gubatan
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kian Keyashian
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Samuel J S Rubin
- Stanford University School of Medicine, Stanford, CA, USA
- Immunology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Jenny Wang
- Stanford University School of Medicine, Stanford, CA, USA
| | | | - Sidhartha Sinha
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
8
|
Shaikh H, Vargas JG, Mokhtari Z, Jarick KJ, Ulbrich M, Mosca JP, Viera EA, Graf C, Le DD, Heinze KG, Büttner-Herold M, Rosenwald A, Pezoldt J, Huehn J, Beilhack A. Mesenteric Lymph Node Transplantation in Mice to Study Immune Responses of the Gastrointestinal Tract. Front Immunol 2021; 12:689896. [PMID: 34381447 PMCID: PMC8352558 DOI: 10.3389/fimmu.2021.689896] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 07/08/2021] [Indexed: 02/02/2023] Open
Abstract
Mesenteric lymph nodes (mLNs) are sentinel sites of enteral immunosurveillance and immune homeostasis. Immune cells from the gastrointestinal tract (GIT) are constantly recruited to the mLNs in steady-state and under inflammatory conditions resulting in the induction of tolerance and immune cells activation, respectively. Surgical dissection and transplantation of lymph nodes (LN) is a technique that has supported seminal work to study LN function and is useful to investigate resident stromal and endothelial cell biology and their cellular interactions in experimental disease models. Here, we provide a detailed protocol of syngeneic mLN transplantation and report assays to analyze effective mLN engraftment in congenic recipients. Transplanted mLNs allow to study T cell activation and proliferation in preclinical mouse models. Donor mLNs proved viable and functional after surgical transplantation and regenerated blood and lymphatic vessels. Immune cells from the host completely colonized the transplanted mLNs within 7-8 weeks after the surgical intervention. After allogeneic hematopoietic cell transplantation (allo-HCT), adoptively transferred allogeneic CD4+ T cells from FVB/N (H-2q) mice homed to the transplanted mLNs in C57BL/6 (H-2b) recipients during the initiation phase of acute graft-versus-host disease (aGvHD). These CD4+ T cells retained full proliferative capacity and upregulated effector and gut homing molecules comparable to those in mLNs from unmanipulated wild-type recipients. Wild type mLNs transplanted into MHCII deficient syngeneic hosts sufficed to activate alloreactive T cells upon allogeneic hematopoietic cell transplantation, even in the absence of MHCII+ CD11c+ myeloid cells. These data support that orthotopically transplanted mLNs maintain physiological functions after transplantation. The technique of LN transplantation can be applied to study migratory and resident cell compartment interactions in mLNs as well as immune reactions from and to the gut under inflammatory and non-inflammatory conditions.
Collapse
Affiliation(s)
- Haroon Shaikh
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Juan Gamboa Vargas
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Zeinab Mokhtari
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Katja J. Jarick
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Maria Ulbrich
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Josefina Peña Mosca
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| | - Estibaliz Arellano Viera
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Caroline Graf
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Duc-Dung Le
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
| | - Katrin G. Heinze
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
- Rudolf Virchow Center, Julius-Maximilians-University Würzburg, Würzburg, Germany
| | - Maike Büttner-Herold
- Department of Nephropathology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Andreas Rosenwald
- Institute of Pathology, Julius-Maximilians-University Würzburg, Würzburg, Germany
- Comprehensive Cancer Centre Mainfranken, Würzburg University Hospital, Würzburg, Germany
| | - Joern Pezoldt
- Laboratory of Systems Biology and Genetics, Institute of Bioengineering, School of Life Sciences, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
| | - Jochen Huehn
- Department of Experimental Immunology, Helmholtz Centre for Infection Research, Braunschweig, Germany
- Cluster of Excellence RESIST (EXC 2155), Hannover Medical School, Hannover, Germany
| | - Andreas Beilhack
- Interdisciplinary Center for Clinical Research (IZKF) Experimental Stem Cell Transplantation Laboratory, Würzburg University Hospital, Würzburg, Germany
- Department of Internal Medicine II, Würzburg University Hospital, Würzburg, Germany
- Graduate School of Life Sciences, Würzburg University, Würzburg, Germany
| |
Collapse
|
9
|
Wiendl M, Becker E, Müller TM, Voskens CJ, Neurath MF, Zundler S. Targeting Immune Cell Trafficking - Insights From Research Models and Implications for Future IBD Therapy. Front Immunol 2021; 12:656452. [PMID: 34017333 PMCID: PMC8129496 DOI: 10.3389/fimmu.2021.656452] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 04/16/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammatory bowel diseases (IBDs), including Crohn's disease (CD) and ulcerative colitis (UC) are multifactorial diseases with still unknown aetiology and an increasing prevalence and incidence worldwide. Despite plentiful therapeutic options for IBDs, the lack or loss of response in certain patients demands the development of further treatments to tackle this unmet medical need. In recent years, the success of the anti-α4β7 antibody vedolizumab highlighted the potential of targeting the homing of immune cells, which is now an important pillar of IBD therapy. Due to its complexity, leukocyte trafficking and the involved molecules offer a largely untapped resource for a plethora of potential therapeutic interventions. In this review, we aim to summarise current and future directions of specifically interfering with immune cell trafficking. We will comment on concepts of homing, retention and recirculation and particularly focus on the role of tissue-derived chemokines. Moreover, we will give an overview of the mode of action of drugs currently in use or still in the pipeline, highlighting their mechanisms and potential to reduce disease burden.
Collapse
Affiliation(s)
- Maximilian Wiendl
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Tanja M. Müller
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Caroline J. Voskens
- Department of Dermatology, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, Deutsches Zentrum Immuntherapie (DZI), University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
10
|
Bilsborough J, Fiorino MF, Henkle BW. Select animal models of colitis and their value in predicting clinical efficacy of biological therapies in ulcerative colitis. Expert Opin Drug Discov 2020; 16:567-577. [PMID: 33245673 DOI: 10.1080/17460441.2021.1851185] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Introduction: Advancing new therapies from discovery to development usually requires proof-of-concept in animal models to justify the costs of continuing the program. While animal models are useful for understanding the mechanism of action (MOA) of a target, limitations of many published colitis models restrict their value to predict clinical efficacy.Areas covered: The authors focused their literature search on published studies of chronic animal models used to evaluate the pre-clinical efficacy of therapeutic molecules subsequently evaluated in clinical trials for UC. The UC therapies evaluated were anti-α4β7, anti-IL13, anti-IL12p40, and anti-IL23p19. The models of chronic colitis evaluating these molecules were: mdra1a-/-, chronic dextran sulfate sodium (DSS), chronic 2,4,6-trinitrobenzene sulfonic acid (TNBS), and the T cell transfer model.Expert opinion: While some models provide insight into target MOA in UC, none is consistently superior in predicting efficacy. Evaluation of multiple models, with varying mechanisms of colitis induction, is needed to understand potential drug efficacy. Additional models of greater complexity, reflecting the disease chronicity/heterogeneity seen in humans, are needed. Although helpful in prioritizing targets, animal models alone will likely not improve outcomes of UC clinical trials. Transformational changes to clinical efficacy will likely only occur when precision medicine approaches are employed.
Collapse
Affiliation(s)
- Janine Bilsborough
- IBD Drug Discovery and Development Unit, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Marie F Fiorino
- IBD Drug Discovery and Development Unit, F. Widjaja Foundation Inflammatory Bowel and Immunbiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Bradley W Henkle
- IBD Drug Discovery and Development Unit, F. Widjaja Foundation Inflammatory Bowel and Immunbiology Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
11
|
Oulghazi S, Wegner SK, Spohn G, Müller N, Harenkamp S, Stenzinger A, Papayannopoulou T, Bonig H. Adaptive Immunity and Pathogenesis of Diabetes: Insights Provided by the α4-Integrin Deficient NOD Mouse. Cells 2020; 9:cells9122597. [PMID: 33291571 PMCID: PMC7761835 DOI: 10.3390/cells9122597] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 11/30/2020] [Accepted: 12/02/2020] [Indexed: 12/12/2022] Open
Abstract
Background: The spontaneously diabetic “non-obese diabetic” (NOD) mouse is a faithful model of human type-1 diabetes (T1D). Methods: Given the pivotal role of α4 integrin (CD49d) in other autoimmune diseases, we generated NOD mice with α4-deficient hematopoiesis (NOD.α4-/-) to study the role of α4 integrin in T1D. Results: NOD.α4-/- mice developed islet-specific T-cells and antibodies, albeit quantitatively less than α4+ counterparts. Nevertheless, NOD.α4-/- mice were completely and life-long protected from diabetes and insulitis. Moreover, transplantation with isogeneic α4-/- bone marrow prevented progression to T1D of pre-diabetic NOD.α4+ mice despite significant pre-existing islet cell injury. Transfer of α4+/CD3+, but not α4+/CD4+ splenocytes from diabetic to NOD.α4-/- mice induced diabetes with short latency. Despite an only modest contribution of adoptively transferred α4+/CD3+ cells to peripheral blood, pancreas-infiltrating T-cells were exclusively graft derived, i.e., α4+. Microbiota of diabetes-resistant NOD.α4-/- and pre-diabetic NOD.α4+ mice were identical. Co- housed diabetic NOD.α4+ mice showed the characteristic diabetic dysbiosis, implying causality of diabetes for dysbiosis. Incidentally, NOD.α4-/- mice were protected from autoimmune sialitis. Conclusion: α4 is a potential target for primary or secondary prevention of T1D.
Collapse
Affiliation(s)
- Salim Oulghazi
- Institute for Transfusion Medicine and Immunohematology, School of Medicine, Goethe University, Sandhofstraße 1, 60528 Frankfurt, Germany or (S.O.); (S.K.W.)
| | - Sarah K. Wegner
- Institute for Transfusion Medicine and Immunohematology, School of Medicine, Goethe University, Sandhofstraße 1, 60528 Frankfurt, Germany or (S.O.); (S.K.W.)
| | - Gabriele Spohn
- Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany; (G.S.); (N.M.); (S.H.)
| | - Nina Müller
- Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany; (G.S.); (N.M.); (S.H.)
| | - Sabine Harenkamp
- Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany; (G.S.); (N.M.); (S.H.)
| | - Albrecht Stenzinger
- Institute for Pathology, University Hospital Heidelberg, Im Neuenheimer Feld 672, 69120 Heidelberg, Germany;
| | - Thalia Papayannopoulou
- Department of Medicine/Division of Hematology, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Halvard Bonig
- Institute for Transfusion Medicine and Immunohematology, School of Medicine, Goethe University, Sandhofstraße 1, 60528 Frankfurt, Germany or (S.O.); (S.K.W.)
- Institute Frankfurt, German Red Cross Blood Service BaWüHe, Sandhofstraße 1, 60528 Frankfurt, Germany; (G.S.); (N.M.); (S.H.)
- Department of Medicine/Division of Hematology, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
- Correspondence: ; Tel.: +49-69-6782177
| |
Collapse
|
12
|
Sofi FA, Bharatam PV. Synthesis of Drugs and Biorelevant N-heterocycles Employing Recent Advances in C-N Bond Formation. CURR ORG CHEM 2020. [DOI: 10.2174/1385272824999200909114144] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
C-N bond formation is a particularly important step in the generation of many
biologically relevant heterocyclic molecules. Several methods have been reported for this
purpose over the past few decades. Well-known named reactions like Ullmann-Goldberg
coupling, Buchwald-Hartwig coupling and Chan-Lam coupling are associated with the C-N
bond formation reactions. Several reviews covering this topic have already been published.
However, no comprehensive review covering the synthesis of drugs/ lead compounds using
the C-N bond formation reactions was reported. In this review, we cover many modern
methods of the C-N bond formation reactions, with special emphasis on metal-free and
green chemistry methods. We also report specific strategies adopted for the synthesis of
drugs, which involve the C-N bond formation reactions. Examples include anti-cancer,
antidepressant, anti-inflammatory, anti-atherosclerotic, anti-histaminic, antibiotics, antibacterial, anti-rheumatic,
antiepileptic and anti-diabetic agents. Many recently developed lead compounds generated using the C-N bond
formation reactions are also covered in this review. Examples include MAP kinase inhibitors, TRKs inhibitors,
Polo-like Kinase inhibitors and MPS1 inhibitors.
Collapse
Affiliation(s)
- Firdoos Ahmad Sofi
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S Nagar 160 062, Punjab, India
| | - Prasad V. Bharatam
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S Nagar 160 062, Punjab, India
| |
Collapse
|
13
|
Holmer AK, Battat R, Dulai PS, Vande Casteele N, Nguyen N, Jain A, Miralles A, Neill J, Le H, Singh S, Rivera-Nieves J, Sandborn WJ, Boland BS. Biomarkers are associated with clinical and endoscopic outcomes with vedolizumab treatment in Crohn's disease. Therap Adv Gastroenterol 2020; 13:1756284820971214. [PMID: 33240396 PMCID: PMC7675888 DOI: 10.1177/1756284820971214] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/14/2020] [Accepted: 10/12/2020] [Indexed: 02/04/2023] Open
Abstract
BACKGROUND Vedolizumab, an α4β7 integrin antagonist, is an effective therapy for Crohn's disease (CD). Biomarkers are needed to guide therapy and predict outcomes. This study evaluated biomarker concentrations and outcomes in patients with CD undergoing vedolizumab treatment. METHODS Sera at weeks 0, 2, 6, 14, and ⩾26 were collected from vedolizumab-treated, refractory CD patients. Concentrations of soluble (s)-Vascular Cell Adhesion Molecule (VCAM)-1, s-Intercellular Cell Adhesion Molecule (ICAM)-1, s-Mucosal Addressin Cell Adhesion Molecule (MAdCAM)-1, and s-α4β7 integrin were evaluated for associations with achieving endoscopic remission. RESULTS A total of 22 patients with CD were included. In all patients, s-MAdCAM-1 decreased significantly and s-α4β7 increased compared with baseline. s-VCAM-1 and s-ICAM-1 changed differentially in patients who achieved remission. At week 6, median s-VCAM-1 (859.6 ng/ml versus 460.3 ng/ml, p = 0.03) and s-ICAM-1 (545.7 ng/ml versus 286.2 ng/ml, p = 0.03) concentrations were higher in patients who achieved endoscopic remission compared with those who did not, and similar differences were observed for s-ICAM-1 concentrations in patients who achieved clinical remission, compared with those who did not (669.1 ng/ml versus 291.0 ng/ml, p = 0.04). Week 14 s-α4β7 concentrations were lower in patients who achieved endoscopic remission, compared with those who did not (7.5 ng/ml versus 17.6 ng/ml, p = 0.020). CONCLUSION In all vedolizumab-treated CD patients, s-MAdCAM-1 decreased significantly and s-α4β7 increased. However, higher concentrations of s-ICAM-1 and s-VCAM-1 at week 6 and lower concentrations of s-α4β7 at week 14 differentiated patients who achieved endoscopic remission. These findings may help identify early predictors of response to vedolizumab treatment in patients with CD. Further validation in less refractory CD patients is needed.
Collapse
Affiliation(s)
| | | | - Parambir S. Dulai
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Niels Vande Casteele
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Nghia Nguyen
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | | | - Ara Miralles
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Jennifer Neill
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Helen Le
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Siddharth Singh
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - Jesus Rivera-Nieves
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | - William J. Sandborn
- Inflammatory Bowel Disease Center, Division of
Gastroenterology, Department of Medicine, University of California, San
Diego, La Jolla, CA, USA
| | | |
Collapse
|
14
|
Therapeutic and Diagnostic Implications of T Cell Scarring in Celiac Disease and Beyond. Trends Mol Med 2019; 25:836-852. [DOI: 10.1016/j.molmed.2019.05.009] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Revised: 05/22/2019] [Accepted: 05/28/2019] [Indexed: 12/13/2022]
|
15
|
T Cell Recruitment to the Intestinal Stem Cell Compartment Drives Immune-Mediated Intestinal Damage after Allogeneic Transplantation. Immunity 2019; 51:90-103.e3. [PMID: 31278057 DOI: 10.1016/j.immuni.2019.06.003] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Revised: 04/01/2019] [Accepted: 06/05/2019] [Indexed: 12/30/2022]
Abstract
The key sites within the gastrointestinal (GI) tract where T cells mediate effector responses and the impact of these responses on intestinal stem cells (ISCs) remain unclear. Using experimental bone marrow transplantation to model immune-mediated GI damage and 3D imaging to analyze T cell localization, we found that the ISC compartment is the primary intestinal site targeted by T cells after transplantation. Recruitment to the crypt base region resulted in direct T cell engagement with the stem cell compartment and loss of crypt base columnar ISCs, which expressed both MHC classes I and II. Vasculature expressing the adhesion molecule MAdCAM-1 clustered near the crypt base, preferentially regulating crypt compartment invasion and ISC reduction without affecting T cell migration to villi. These findings indicate that allogeneic T cells rapidly access the stem cell niche after transplantation, and this targeted recruitment to the stem cell compartment results in ISC loss during immune-mediated GI damage.
Collapse
|
16
|
Targeting immune cell circuits and trafficking in inflammatory bowel disease. Nat Immunol 2019; 20:970-979. [PMID: 31235952 DOI: 10.1038/s41590-019-0415-0] [Citation(s) in RCA: 437] [Impact Index Per Article: 72.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 05/01/2019] [Indexed: 12/20/2022]
Abstract
Inflammatory bowel diseases (IBDs) such as Crohn's disease and ulcerative colitis are characterized by uncontrolled activation of intestinal immune cells in a genetically susceptible host. Due to the progressive and destructive nature of the inflammatory process in IBD, complications such as fibrosis, stenosis or cancer are frequently observed, which highlights the need for effective anti-inflammatory therapy. Studies have identified altered trafficking of immune cells and pathogenic immune cell circuits as crucial drivers of mucosal inflammation and tissue destruction in IBD. A defective gut barrier and microbial dysbiosis induce such accumulation and local activation of immune cells, which results in a pro-inflammatory cytokine loop that overrides anti-inflammatory signals and causes chronic intestinal inflammation. This Review discusses pathogenic cytokine responses of immune cells as well as immune cell trafficking as a rational basis for new translational therapies in IBD.
Collapse
|
17
|
Dotan I, Allez M, Danese S, Keir M, Tole S, McBride J. The role of integrins in the pathogenesis of inflammatory bowel disease: Approved and investigational anti-integrin therapies. Med Res Rev 2019; 40:245-262. [PMID: 31215680 PMCID: PMC6973243 DOI: 10.1002/med.21601] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 04/12/2019] [Accepted: 05/13/2019] [Indexed: 12/12/2022]
Abstract
Inflammatory bowel disease (IBD) is characterized by uncontrolled inflammation in the gastrointestinal tract. The underlying pathobiology of IBD includes an increase in infiltrating gut-homing lymphocytes. Although lymphocyte homing is typically a tightly regulated and stepwise process involving multiple integrins and adhesion molecules expressed on endothelial cells, the distinct roles of integrin-expressing immune cells is not fully understood in the pathology of IBD. In this review, we detail the involvement of integrins expressed on specific lymphocyte subsets in the pathogenesis of IBD and discuss the current status of approved and investigational integrin-targeted therapies.
Collapse
Affiliation(s)
- Iris Dotan
- Division of Gastroenterology, Rabin Medical Center, Petah Tikva, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Matthieu Allez
- Department of Gastroenterology, Hôpital Saint-Louis, AP-HP, INSERM U1160, University Denis Diderot, Paris, France
| | - Silvio Danese
- Gastrointestinal Immunopathology Laboratory and IBD Unit, Humanitas Clinical and Research Center, Milan, Italy
| | - Mary Keir
- Department of Research and Early Development, Genentech, South San Francisco, California
| | - Swati Tole
- Department of Product Development, Genentech, South San Francisco, California
| | - Jacqueline McBride
- Department of Research and Early Development, Genentech, South San Francisco, California
| |
Collapse
|
18
|
Battat R, Dulai PS, Jairath V, Vande Casteele N. A product review of vedolizumab in inflammatory bowel disease. Hum Vaccin Immunother 2019; 15:2482-2490. [PMID: 30897022 DOI: 10.1080/21645515.2019.1591139] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Vedolizumab is a monoclonal antibody to the α4β7 integrin that selectively reduces intestinal lymphocyte trafficking, thereby providing a safe and effective treatment option for patients with inflammatory bowel disease (IBD). This product review outlines the unique mechanism of vedolizumab in addition to efficacy, safety, pharmacokinetic and pharmacodynamic data from clinical trials, observational studies and meta-analyses. Vedolizumab has been shown to be effective as a first- or second-line induction and maintenance therapy in both ulcerative colitis (UC) and Crohn's disease (CD). Prolonged induction therapy may increase efficacy, particularly in tumor necrosis factor-alpha-exposed CD patients. To date, no drug-specific safety signals have been identified. In addition to the presence of an apparent exposure-response relationship, vedolizumab has demonstrated consistent pharmacodynamic effects on α4β7, mucosal vascular addressin cell adhesion molecule 1 and other cell adhesion molecules. Future efforts should focus on identifying predictive biomarkers capable of guiding personalized IBD treatment with vedolizumab.
Collapse
Affiliation(s)
- Robert Battat
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, University of California San Diego , La Jolla , CA , USA.,Robarts Clinical Trials Inc ., London , ON , Canada
| | - Parambir S Dulai
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, University of California San Diego , La Jolla , CA , USA.,Robarts Clinical Trials Inc ., London , ON , Canada
| | - Vipul Jairath
- Robarts Clinical Trials Inc ., London , ON , Canada.,Department of Medicine, University of Western Ontario , London , ON , Canada.,Department of Epidemiology and Biostatistics, University of Western Ontario , London , ON , Canada
| | - Niels Vande Casteele
- Inflammatory Bowel Disease Center, Division of Gastroenterology, Department of Medicine, University of California San Diego , La Jolla , CA , USA.,Robarts Clinical Trials Inc ., London , ON , Canada
| |
Collapse
|
19
|
Biswas S, Bryant RV, Travis S. Interfering with leukocyte trafficking in Crohn's disease. Best Pract Res Clin Gastroenterol 2019; 38-39:101617. [PMID: 31327399 DOI: 10.1016/j.bpg.2019.05.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/23/2018] [Accepted: 05/23/2019] [Indexed: 01/31/2023]
Abstract
The discovery of gut-specific leukocytes and the ability to modulate their function has been a groundbreaking development in the treatment of inflammatory bowel disease. Drugs target the interaction between lymphocytes and endothelial cells via integrins and their ligand cellular adhesion molecules. Safety, efficacy and sustainability of effect are key to this drug class, notwithstanding the association of natalizumab with fatal polyoma virus infection. Vedolizumab (2014) now licensed for the treatment of Crohn's disease around the world provides gut-specific immunosuppression. Targets for modulators of leukocyte trafficking include (examples in brackets) ICAM-1 (alicaforsen, efalizumab); MAdCAM-1 (PF-00547 659); α4 and related receptors (abrilumab, etrolizumab, natalizumab, vedolizumab); chemokine receptor CCR9 (vercirnon); and sphingosine 1-phosphate receptors (etrasimod, fingolimod, ozanimod). Oral and subcutaneous therapies are in development. The safety, efficacy and practice points of licensed drugs are discussed, in addition to initial results from therapeutic trials.
Collapse
Affiliation(s)
- Sujata Biswas
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| | - Robert V Bryant
- IBD Service, Department of Gastroenterology, The Queen Elizabeth Hospital, Woodville South, Adelaide, South Australia, 5011, Australia.
| | - Simon Travis
- Translational Gastroenterology Unit, NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, OX3 9DU, UK.
| |
Collapse
|
20
|
Battat R, Dulai PS, Vande Casteele N, Evans E, Hester KD, Webster E, Jain A, Proudfoot JA, Mairalles A, Neill J, Singh S, Chang JT, Rivera-Nieves J, Sandborn WJ, Boland BS. Biomarkers Are Associated With Clinical and Endoscopic Outcomes With Vedolizumab Treatment in Ulcerative Colitis. Inflamm Bowel Dis 2019; 25:410-420. [PMID: 30295781 PMCID: PMC6327228 DOI: 10.1093/ibd/izy307] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Indexed: 02/07/2023]
Abstract
Background Vedolizumab inhibits α4β7-mediated lymphocyte trafficking and is effective in ulcerative colitis (UC). This study evaluated drug and biomarker concentrations and patient outcomes during vedolizumab treatment in UC. Methods Prospectively scored maintenance clinical (26.5 weeks; interquartile range [IQR], 16.3-37.0 weeks) and endoscopic (23.5 weeks; IQR, 16.8-35.6 weeks) outcomes were compared with serum vedolizumab concentrations, antivedolizumab antibodies, and serum biomarkers at baseline and weeks 2, 6, 14, and 26. A linear mixed-effects model compared biomarker trajectories over time between clinical and endoscopic remitters and nonremitters. Results Thirty-two patients were included. Soluble (s)-tumor necrosis factor (TNF)-α, s-α4β7, s-mucosal addressin cell adhesion molecule (s-MAdCAM-1), and s-amyloid A (s-AA) significantly changed with treatment. A linear mixed-effects model demonstrated that s-α4β7 (P = 0.044) increased and s-MAdCAM-1 (P = 0.006) and s-vascular cell adhesion molecule-1 (s-VCAM-1, P = 0.001) decreased more rapidly in patients achieving clinical remission in maintenance. S-MAdCAM-1 (P = 0.005), s-intracellular adhesion molecule-1 (ICAM-1; P = 0.014), s-VCAM-1 (P < 0.001), and s-TNF (P = 0.052) decreased more rapidly in endoscopic remitters. In clinical remitters, higher week 14 (20.3 ng/mL vs 6.0 ng/mL; P = 0.013) and week 26 (14.1 ng/mL vs 8.6 ng/mL; P = 0.05) s-α4β7 were observed. In endoscopic remitters, week 2 (6.7 pg/mL vs 17.8 pg/mL; P = 0.038) and week 6 (3.9 pg/mL vs 15.6 pg/mL; P = 0.005) s-TNF and week 14 s-VCAM (589.1 ng/mL vs 746.0 ng/mL; P = 0.05) were lower. Conclusion Serum biomarkers were associated with outcomes in vedolizumab-treated UC patients. s-α4β7 increased, whereas s-MAdCAM-1, s-VCAM-1, s-ICAM-1, and s-TNF decreased more rapidly in remitters. At individual time points, induction s-TNF and maintenance s-VCAM-1 concentrations were lower, whereas maintenance s-α4β7 concentrations were higher in remitters.
Collapse
Affiliation(s)
- Robert Battat
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Parambir S Dulai
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Niels Vande Casteele
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Elisabeth Evans
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | | | | | - Anjali Jain
- Prometheus Laboratories Inc., San Diego, California
| | - James A Proudfoot
- Clinical and Translational Research Institute, University of California, San Diego, La Jolla, California
| | - Ara Mairalles
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Jennifer Neill
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Siddharth Singh
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - John T Chang
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Jesus Rivera-Nieves
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - William J Sandborn
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| | - Brigid S Boland
- Division of Gastroenterology, Department of Medicine, Inflammatory Bowel Disease Center, University of California, San Diego, La Jolla, California
| |
Collapse
|
21
|
Lupsa N, Érsek B, Horváth A, Bencsik A, Lajkó E, Silló P, Oszvald Á, Wiener Z, Reményi P, Mikala G, Masszi T, Buzás EI, Pós Z. Skin-homing CD8 + T cells preferentially express GPI-anchored peptidase inhibitor 16, an inhibitor of cathepsin K. Eur J Immunol 2018; 48:1944-1957. [PMID: 30365157 DOI: 10.1002/eji.201847552] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2018] [Revised: 09/07/2018] [Accepted: 10/16/2018] [Indexed: 12/29/2022]
Abstract
This study sought to identify novel CD8+ T cell homing markers by studying acute graft versus host disease (aGvHD), typically involving increased T cell homing to the skin and gut. FACS-sorted skin-homing (CD8β+ /CLA+ ), gut-homing (CD8β+ /integrinβ7+ ), and reference (CD8β+ /CLA- /integrinβ7- ) T cells were compared in patients affected by cutaneous and/or gastrointestinal aGVHD. Microarray analysis, qPCR, and flow cytometry revealed increased expression of peptidase inhibitor 16 (PI16) in skin-homing CD8+ T cells. Robust association of PI16 with skin homing was confirmed in all types of aGvHD and in healthy controls, too. PI16 was not observed on CLA+ leukocytes other than T cells. Induction of PI16 expression on skin-homing T cells occurred independently of vitamin D3. Among skin-homing T cells, PI16 expression was most pronounced in memory-like CD45RO+ /CD127+ /CD25+ /CD69- /granzyme B- cells. PI16 was confined to the plasma membrane, was GPI-anchored, and was lost upon restimulation of memory CD8+ T cells. Loss of PI16 occurred by downregulation of PI16 transcription, and not by Phospholipase C (PLC)- or Angiotensin-converting enzyme (ACE)-mediated shedding, or by protein recycling. Inhibitor screening and pull-down experiments confirmed that PI16 inhibits cathepsin K, but may not bind to other skin proteases. These data link PI16 to skin-homing CD8+ T cells, and raise the possibility that PI16 may regulate cutaneous cathepsin K.
Collapse
Affiliation(s)
- Nikolett Lupsa
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary.,Hungarian Academy of Sciences, Semmelweis University Immunoproteogenomics Extracellular Vesicle Research Group, Budapest, Hungary
| | - Barbara Érsek
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary.,Office for Research Groups Attached to Universities and Other Institutions, Hungarian Academy of Sciences, Budapest, Hungary
| | - Andor Horváth
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - András Bencsik
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Eszter Lajkó
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Pálma Silló
- Department of Dermatology, Venereology and Dermatooncology, Semmelweis University, Budapest, Hungary
| | - Ádám Oszvald
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Zoltán Wiener
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| | - Péter Reményi
- Department of Hematology and Stem Cell Transplantation, St. Istvan and Saint Laszlo Hospital, Budapest, Hungary
| | - Gábor Mikala
- Department of Hematology and Stem Cell Transplantation, St. Istvan and Saint Laszlo Hospital, Budapest, Hungary
| | - Tamás Masszi
- Department of Hematology and Stem Cell Transplantation, St. Istvan and Saint Laszlo Hospital, Budapest, Hungary.,3rd Department of Internal Medicine, Semmelweis University, Budapest, Hungary
| | - Edit I Buzás
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary.,Hungarian Academy of Sciences, Semmelweis University Immunoproteogenomics Extracellular Vesicle Research Group, Budapest, Hungary
| | - Zoltán Pós
- Department of Genetics, Cell and Immunobiology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
22
|
Lamb CA, O'Byrne S, Keir ME, Butcher EC. Gut-Selective Integrin-Targeted Therapies for Inflammatory Bowel Disease. J Crohns Colitis 2018; 12:S653-S668. [PMID: 29767705 DOI: 10.1093/ecco-jcc/jjy060] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Integrins are cell surface receptors with bidirectional signalling capabilities that can bind to adhesion molecules in order to mediate homing of leukocytes to peripheral tissues. Gut-selective leukocyte homing is facilitated by interactions between α4β7 and its ligand, mucosal addressin cellular adhesion molecule-1 [MAdCAM-1], while retention of lymphocytes in mucosal tissues is mediated by αEβ7 binding to its ligand E-cadherin. Therapies targeting gut-selective trafficking have shown efficacy in inflammatory bowel disease [IBD], confirming the importance of leukocyte trafficking in disease pathobiology. This review will provide an overview of integrin structure, function and signalling, and highlight the role that these molecules play in leukocyte homing and retention. Anti-integrin therapeutics, including gut-selective antibodies against the β7 integrin subunit [etrolizumab] and the α4β7 integrin heterodimer [vedolizumab and abrilumab], and the non-gut selective anti-α4 integrin [natalizumab], will be discussed, as well as novel targeting approaches using small molecules.
Collapse
Affiliation(s)
- Christopher A Lamb
- Institute of Cellular Medicine, Newcastle University, Newcastle upon Tyne, UK.,Department of Gastroenterology, Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - Sharon O'Byrne
- Global Medical Affairs, Takeda Pharmaceuticals International AG, Zurich, Switzerland
| | - Mary E Keir
- Genentech Research & Early Development, South San Francisco, CA, USA
| | - Eugene C Butcher
- Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA.,Veterans Affairs Palo Alto Health Care System and The Palo Alto Veterans Institute for Research, Palo Alto, CA, USA
| |
Collapse
|
23
|
Panés J, Salas A. Past, Present and Future of Therapeutic Interventions Targeting Leukocyte Trafficking in Inflammatory Bowel Disease. J Crohns Colitis 2018; 12:S633-S640. [PMID: 30137311 DOI: 10.1093/ecco-jcc/jjy011] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Studies in the 1990s using animal models of intestinal inflammation delineated the crucial molecules involved in leukocyte attraction and retention to the inflamed gut and associated lymphoid tissues. The first drug targeting leukocyte trafficking tested in inflammatory bowel diseases was the anti-ICAM-1 antisense oligonucleotide alicaforsen, showing only modest efficacy. Subsequently, the anti-α4 monoclonal antibody natalizumab proved efficacious for induction and maintenance of remission in Crohn's disease, but was associated with progressive multifocal leukoencephalopathy due to its ability to interfere with both α4β1 and α4β7 function. Later developments in this area took advantage of the fairly selective expression of MAdCAM-1 in the digestive organs, showing that vedolizumab, a more specific monoclonal antibody selectively blocking MAdCAM-1 binding to integrin α4β7, was efficacious for induction and maintenance of remission in ulcerative colitis and Crohn's disease, and it was not associated with neurological complications. Currently, other drugs targeting the β7 subunit, immunoglobulin superfamily molecules expressed on the endothelium, as well as blockade of lymphocyte recirculation in lymph nodes through modulation of sphingosine 1-phosphate receptors are under development. The potential use and risks of combined anti-trafficking therapy will be examined in this review.
Collapse
Affiliation(s)
- Julián Panés
- Department of Gastroenterology, Hospital Clínic de Barcelona, Barcelona, Spain.,Institut d'investigacions Biomèdiques August Pi i Sunyer, CIBERehd, Barcelona, Spain
| | - Azucena Salas
- Institut d'investigacions Biomèdiques August Pi i Sunyer, CIBERehd, Barcelona, Spain
| |
Collapse
|
24
|
Nelson SML, Nguyen TM, McDonald JWD, MacDonald JK. Natalizumab for induction of remission in Crohn's disease. Cochrane Database Syst Rev 2018; 8:CD006097. [PMID: 30068022 PMCID: PMC6513248 DOI: 10.1002/14651858.cd006097.pub3] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
BACKGROUND This systematic review update summarizes the current evidence on the use of natalizumab for induction of remission in Crohn's disease (CD). OBJECTIVES To determine the efficacy and safety of natalizumab for induction of remission in CD. SEARCH METHODS We searched MEDLINE, Embase, CENTRAL, the Cochrane IBD Group Specialized Trials Register, and clinicaltrials.gov from inception to 10 May 2018. SELECTION CRITERIA We included randomized controlled trials (RCTs) comparing natalizumab to a placebo or control therapy for induction of remission in CD. DATA COLLECTION AND ANALYSIS Two authors independently screened studies, extracted data and assessed methodological quality using the Cochrane risk of bias tool. The primary outcome was failure to enter clinical remission. Secondary outcomes included clinical response, mean change in Crohn's Disease Activity Index (CDAI), adverse events (AEs), withdrawal due to AEs and serious AEs. For dichotomous outcomes, we calculated the risk ratio (RR) and 95% confidence interval (95% CI). For continuous outcomes we calculated the mean difference (MD) and 95% CI. Data were pooled for meta-analysis when the interventions, patient groups and outcomes were sufficiently similar (determined by consensus). We used GRADE to assess the overall quality of the evidence. MAIN RESULTS A total of five RCTs (1771 participants) were included. Four studies (1692 participants) compared one, two or three infusions of natalizumab (300 mg or 3 mg/kg or 6mg/kg) to placebo. One study (79 participants) compared three infusions of natalizumab (300 mg) and infliximab (5 mg/kg) to infliximab and placebo. Four studies were rated as low risk of bias. One study was rated as unclear risk of bias for selective reporting.One, two and three infusions of natalizumab were superior to placebo for induction of remission and clinical response. Infusions were administered at weeks zero, four and eight. After one infusion, 76% (849/1117) of natalizumab participants failed to enter remission at 4 weeks compared to 83% (411/494) of placebo participants (RR 0.91, 95% CI 0.86 to 0.96, 3 studies, GRADE high quality). At 4 weeks, the RR for clinical response was 0.78 (95% CI 0.66 to 0.92, 3 studies, 1611 participants, GRADE moderate quality). After two infusions, after 8 weeks, 66% (693/1049) of natalizumab participants failed to enter remission compared to 77% (382/494) of placebo participants (RR 0.85, 95% CI 0.76 to 0.95; 3 studies, GRADE moderate quality). At 8 weeks, the RR for clinical response was 0.73 (95% CI 0.58 to 0.91, 3 studies, 1543 participants, GRADE low quality). After three infusions, at 12 weeks, 61% (596/983) of natalizumab participants failed to enter remission compared to 73% (313/431) of placebo participants (RR 0.85, 95% CI 0.78 to 0.92, 2 studies, GRADE high quality). At 12 weeks, the RR for clinical response was 0.76 (95% CI 0.67 to 0.86, 2 studies, 1414 participants, GRADE high quality). One study (507 participants) reported on change in CADI from baseline. Natalizumab participants had a larger drop in mean CDAI scores than placebo participants at 4, 8 and 12 weeks.The rates of AEs, withdrawals due to AEs and serious AEs were similar across groups at 4, 8 and 12 weeks. After one infusion, 74% (50/68) of natalizumab participants experienced an AE compared to 81% (51/63) of placebo participants (RR 0.91, 95% CI 0.75 to 1.09, GRADE moderate quality). Withdrawal due to an AE occurred in 1% (1/68) of natalizumab participants and 3% of placebo participants (RR 0.46, 95% CI 0.04 to 4.98, GRADE low quality). SAEs occurred in 10% (7/68) of natalizumab participants compared to 11% (7/63) of placebo participants (RR 0.93, 95% CI 0.34 to 2.49, GRADE low quality). After two infusions, 86% (57/66) of natalizumab participants experienced an AE compared to 81% (51/63) of placebo participants (RR 1.07, 95% CI 0.92 to 1.24, GRADE moderate quality). Withdrawal due to an AE occurred in 3% (2/66) natalizumab participants compared to 3% (2/63) placebo participants (RR 0.95, 95% CI 0.14 to 6.57, GRADE low quality). SAEs occurred in 9% (6/66) of natalizumab participants and 11% (7/63) of placebo participants (RR 0.82, 95% CI 0.29 to 2.30, GRADE low quality). After three infusions, 86% (848/984) of natalizumab participants experienced an AE compared to 83% (359/431) placebo participants (RR 1.03, 95% CI 0.98 to 1.08, GRADE high quality). Withdrawals due to AEs occurred in 8% (82/984) of natalizumab participants compared to 10% (45/431) of placebo participants (RR 0.86, 95% CI 0.59 to 1.26, GRADE moderate quality). SAEs occurred in 7% (65/983) of natalizumab participants and 8% (36/431) of placebo participants (RR 0.76. 95% CI 0.37 to 1.56, GRADE low quality). Adverse events included headache, nausea, nasopharyngitis, abdominal pain, fatigue, vomiting, and exacerbation of CD.The study comparing combination therapy with natalizumab and infliximab to infliximab and placebo demonstrated similar remission rates at 10 weeks. Sixty-four per cent (33/52) of participants assigned to natalizumab and infliximab failed to achieve remission compared to 70% (19/27) assigned to placebo and infliximab (RR 0.90, 95% CI 0.65 to 1.24, GRADE moderate quality). The rates of AEs (moderate quality evidence), withdrawals due to AEs (low quality evidence) and serious AEs (low quality evidence) were similar across groups at 10 weeks. Adverse events included headache, exacerbation of CD, nausea, and nasopharyngitis.Natalizumab is associated with the development of progressive multifocal leukoencephalopathy (PML) resulting in some patient deaths. There are currently no tests which can reliably predict those at risk of developing PML. AUTHORS' CONCLUSIONS High quality data suggest that natalizumab is effective for induction of clinical remission and response in some patients with moderately to severely active CD. However, none of the included studies had the power to detect rare but serious adverse events such as PML. Due to the association with PML, and the availability of alternative agents that are not associated with PML, natalizumab is not likely to be used in patients who fail currently available medical therapy. The use of natalizumab in select patients (e.g. patients allergic to different biologics) needs to be carefully considered against the potential risk of developing PML. Futher studies of natalizumab are not likely to be done.
Collapse
Affiliation(s)
- Seana ML Nelson
- University of Western OntarioDepartment of MedicineLondonCanada
| | - Tran M Nguyen
- Robarts Clinical TrialsCochrane IBD Group100 Dundas Street, Suite 200LondonONCanada
| | - John WD McDonald
- Robarts Clinical TrialsCochrane IBD Group100 Dundas Street, Suite 200LondonONCanada
| | - John K MacDonald
- Robarts Clinical TrialsCochrane IBD Group100 Dundas Street, Suite 200LondonONCanada
| | | |
Collapse
|
25
|
Rath T, Billmeier U, Ferrazzi F, Vieth M, Ekici A, Neurath MF, Atreya R. Effects of Anti-Integrin Treatment With Vedolizumab on Immune Pathways and Cytokines in Inflammatory Bowel Diseases. Front Immunol 2018; 9:1700. [PMID: 30131801 PMCID: PMC6090141 DOI: 10.3389/fimmu.2018.01700] [Citation(s) in RCA: 42] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/10/2018] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND AIMS Despite proven clinical efficacy of vedolizumab (VDZ) for inducing and maintaining remission in patients with Crohn's disease (CD) and ulcerative colitis (UC), subgroups of patients have no therapeutic benefit from anti-α4β7 integrin therapy with VDZ. Within this study, we aimed to identify genetic, cellular, and immunological mechanisms that define response and failure to VDZ treatment. METHODS Intestinal RNA sequencing was performed in UC and CD patients before and at week 14 of VDZ therapy. α4β7 expression on peripheral and mucosal immune cells was assessed by flow cytometry and immunohistochemistry. Cellular modes of VDZ-mediated action were analyzed ex vivo and in VDZ-treated inflammatory bowel disease patients. RESULTS Transcriptome analysis showed an impairment of signaling cascades associated with adhesion, diapedesis, and migration of granulocytes and agranulocytes upon VDZ therapy. In non-remitters to VDZ therapy, a tissue destructive and leukocyte-mediated inflammatory activity with activation of TNF-dependent pathways was present, all of which were inhibited in remitters to VDZ. Clinical remission was associated with a significant reduction of α4β7 expression on Th2 and Th17 polarized mucosal CD4+ T cells at week 14 of VDZ therapy and with significantly higher numbers of α4β7-expressing mucosal cells prior to the initiation of VDZ therapy compared with non-remitters. CONCLUSION Intestinal α4β7 expression prior to VDZ therapy might represent a biomarker that predicts therapeutic response to subsequent VDZ treatment. Due to high activation of TNF signaling in VDZ non-remitters, anti-TNF treatment might represent a promising therapeutic strategy in VDZ refractory patients.
Collapse
Affiliation(s)
- Timo Rath
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Ulrike Billmeier
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth, Bayreuth, Germany
| | - Arif Ekici
- Institute of Human Genetics, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Markus F. Neurath
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, Division of Gastroenterology, Pneumology and Endocrinology, Ludwig Demling Endoscopy Center of Excellence, University of Erlangen-Nuremberg, Erlangen, Germany
| |
Collapse
|
26
|
Kebir H, Carmant L, Fontaine F, Béland K, Bosoi CM, Sanon NT, Alvarez JI, Desgent S, Pittet CL, Hébert D, Langlois MJ, Rébillard RM, Nguyen DK, Cieuta-Walti C, Holmes GL, Goodkin HP, Mytinger JR, Connolly MB, Prat A, Haddad E. Humanized mouse model of Rasmussen's encephalitis supports the immune-mediated hypothesis. J Clin Invest 2018; 128:2000-2009. [PMID: 29629902 DOI: 10.1172/jci97098] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2017] [Accepted: 02/13/2018] [Indexed: 12/21/2022] Open
Abstract
Rasmussen's encephalitis (RE) is a chronic inflammatory brain disorder that causes frequent seizures and unilateral hemispheric atrophy with progressive neurological deficits. Hemispherectomy remains the only treatment that leads to seizure freedom for this refractory epileptic syndrome. The absence of an animal model of disease has been a major obstacle hampering the development of effective therapies. Here, we describe an experimental mouse model that shares several clinical and pathological features with the human disease. Immunodeficient mice injected with peripheral blood mononuclear cells from RE patients and monitored by video electroencephalography developed severe seizures of cortical origin and showed intense astrogliosis and accumulation of human IFN-γ- and granzyme B-expressing T lymphocytes in the brain compared with mice injected with immune cells from control subjects. We also provide evidence for the efficacy of α4 integrin blockade, an approved therapy for the treatment of multiple sclerosis and Crohn's disease, in reducing inflammatory markers associated with RE in the CNS. This model holds promise as a valuable tool for understanding the pathology of RE and for developing patient-tailored experimental therapeutics.
Collapse
Affiliation(s)
- Hania Kebir
- Department of Microbiology, Infectious Diseases, and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Lionel Carmant
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada.,Department of Pediatrics, and.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - François Fontaine
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada.,Department of Pediatrics, and
| | - Kathie Béland
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Ciprian M Bosoi
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Nathalie T Sanon
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Jorge I Alvarez
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sébastien Desgent
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Camille L Pittet
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - David Hébert
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | - Marie-Josée Langlois
- Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada
| | | | - Dang K Nguyen
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Cécile Cieuta-Walti
- Centre Hospitalier Universitaire de Sherbrooke, Department of Pediatrics, Université de Sherbrooke, Shebrooke, Quebec, Canada
| | - Gregory L Holmes
- Department of Neurological Sciences, Larner College of Medicine, University of Vermont, Burlington, Vermont, USA
| | - Howard P Goodkin
- Fontaine Research Park, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| | - John R Mytinger
- Fontaine Research Park, Department of Pediatrics, University of Virginia, Charlottesville, Virginia, USA
| | - Mary B Connolly
- B.C. Children's Hospital, Division of Neurology, Department of Pediatrics, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alexandre Prat
- Centre de Recherche du Centre Hospitalier de l'Université de Montréal (CRCHUM), Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Department of Neurosciences, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada
| | - Elie Haddad
- Department of Microbiology, Infectious Diseases, and Immunology, Faculty of Medicine, Université de Montréal, Montreal, Quebec, Canada.,Centre de Recherche du Centre Hospitalier Universitaire Sainte-Justine (CHU Sainte-Justine), Montreal, Quebec, Canada.,Department of Pediatrics, and
| |
Collapse
|
27
|
Pandolfi F, Franza L, Altamura S, Mandolini C, Cianci R, Ansari A, Kurnick JT. Integrins: Integrating the Biology and Therapy of Cell-cell Interactions. Clin Ther 2017; 39:2420-2436. [PMID: 29203050 DOI: 10.1016/j.clinthera.2017.11.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2017] [Revised: 10/29/2017] [Accepted: 11/07/2017] [Indexed: 12/12/2022]
Abstract
PURPOSE Although the role of integrins has been described in a variety of diseases, these roles seem to be distinct. To date, no study has attempted to provide links to the various pathways by which such integrins can be involved in these diverse disease settings. The purpose of this review was to address this gap in our knowledge with the hypothesis that there is, in fact, a common pathway by which integrins may function. METHODS This article provides an in-depth perspective on the discovery, development, and design of therapeutics that modulate cellular function by targeting integrin:ligand interactions by reviewing the literature on this subject; the review included the most recent results of clinical and subclinical studies. A MEDLINE search was conducted for articles pertaining to the various issues related to integrins, and the most relevant articles are discussed (ie, not only those published in journals with a higher impact factor). FINDINGS It seems that the ligation of the integrins with their cognate ligands plays a major role in translating membrane dialogue into biological function. In addition, they also seem to play a major regulatory role that can enhance or inhibit biological function depending on the context within which such receptor:ligand interactions occur and the organ and tissues at which interactions occurs and is manipulated. Those studies that used statistical analyses have been included where appropriate. IMPLICATIONS Our findings show that anti-integrin treatment has the potential to become a valid coadjuvant in the treatment of several diseases including cancer, inflammatory diseases, HIv infection and cardiovascular diseases.
Collapse
Affiliation(s)
- Franco Pandolfi
- Institute of Internal Medicine, Catholic University, Rome, Italy.
| | - Laura Franza
- Institute of Internal Medicine, Catholic University, Rome, Italy
| | - Simona Altamura
- Institute of Internal Medicine, Catholic University, Rome, Italy
| | | | - Rossella Cianci
- Institute of Internal Medicine, Catholic University, Rome, Italy
| | - Aftab Ansari
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, Georgia
| | - James T Kurnick
- CytoCure LLC, Beverly, Massachusetts; Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
28
|
Srivastava A, Gupta J, Kumar S, Kumar A. Gut biofilm forming bacteria in inflammatory bowel disease. Microb Pathog 2017; 112:5-14. [PMID: 28942174 DOI: 10.1016/j.micpath.2017.09.041] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2017] [Revised: 09/15/2017] [Accepted: 09/18/2017] [Indexed: 12/18/2022]
Abstract
Inflammatory bowel disease (IBD) symbolizes a group of intestinal disorders in which prolonged inflammation occur in the digestive tract (esophagus, large intestine, small intestine mouth, stomach). Both genetic and environmental factors (infections, stress, diet) are involved in the development of IBD. As we know that bacteria are found in the intestinal mucosa of human and clinical observations revealed bacterial biofilms associated with patients of IBD. Various factors and microbes are found to play an essential role in biofilm formation and mucosal colonization during IBD. Biofilm formation in the digestive tract is dependent on an extracellular matrix synthesized by the bacteria and it has an adverse effect on the immune response of the host. There is no satisfactory and safe treatment option for IBD. Therefore, the current research aims to disrupt biofilm in IBD and concentrates predominantly on improving the drug. Here, we review the literature on bacterial biofilm and IBD to gather new knowledge on the current understanding of biofilm formation in IBD, host immune deregulation and dysbiosis in IBD, molecular mechanism, bacteria involved in biofilm formation, current and future regimen. It is urgently required to plan new ways to control and eradicate bacteria in biofilms that will open up novel diagnostic and therapeutic avenues for IBD. This article includes the mechanism of signaling molecules with respect to the biofilm-related genes as well as the diagnostic methods and new technologies involved in the treatment of IBD.
Collapse
Affiliation(s)
- Ankita Srivastava
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Vill: Hadauri, Post: Tindola, Dist: Barabanki 225003, Uttar Pradesh, India
| | - Jaya Gupta
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Vill: Hadauri, Post: Tindola, Dist: Barabanki 225003, Uttar Pradesh, India
| | - Sunil Kumar
- Faculty of Biotechnology, Institute of Biosciences and Technology, Shri Ramswaroop Memorial University, Lucknow-Deva Road, Vill: Hadauri, Post: Tindola, Dist: Barabanki 225003, Uttar Pradesh, India.
| | - Awanish Kumar
- Department of Biotechnology, National Institute of Technology, Raipur 492010, Chhattisgarh, India.
| |
Collapse
|
29
|
Zundler S, Becker E, Weidinger C, Siegmund B. Anti-Adhesion Therapies in Inflammatory Bowel Disease-Molecular and Clinical Aspects. Front Immunol 2017; 8:891. [PMID: 28804488 PMCID: PMC5532375 DOI: 10.3389/fimmu.2017.00891] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2017] [Accepted: 07/12/2017] [Indexed: 12/22/2022] Open
Abstract
The number of biologicals for the therapy of immunologically mediated diseases is constantly growing. In contrast to other agents that were previously introduced in rheumatologic or dermatologic diseases and only later adopted for the treatment of inflammatory bowel diseases (IBDs), the field of IBD was ground breaking for the concept of anti-adhesion blockade. Anti-adhesion antibodies selectively target integrins controlling cell homing to the intestine, which leads to reduction of inflammatory infiltration to the gut in chronic intestinal inflammation. Currently, the anti-α4β7-antibody vedolizumab is successfully used for both Crohn's disease and ulcerative colitis worldwide. In this mini-review, we will summarize the fundamental basis of intestinal T cell homing and explain the molecular groundwork underlying current and potential future anti-adhesion therapies. Finally, we will comment on noteworthy clinical aspects of anti-adhesion therapy and give an outlook to the future of anti-integrin antibodies and inhibitors.
Collapse
Affiliation(s)
- Sebastian Zundler
- Department of Medicine 1, University of Erlangen-Nuremberg, Translational Research Center, Erlangen, Germany
| | - Emily Becker
- Department of Medicine 1, University of Erlangen-Nuremberg, Translational Research Center, Erlangen, Germany
| | - Carl Weidinger
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-University Medicine, Berlin, Germany.,Berlin Institute of Health, Berlin, Germany
| | - Britta Siegmund
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Charité-University Medicine, Berlin, Germany
| |
Collapse
|
30
|
Drescher HK, Schippers A, Clahsen T, Sahin H, Noels H, Hornef M, Wagner N, Trautwein C, Streetz KL, Kroy DC. β 7-Integrin and MAdCAM-1 play opposing roles during the development of non-alcoholic steatohepatitis. J Hepatol 2017; 66:1251-1264. [PMID: 28192190 DOI: 10.1016/j.jhep.2017.02.001] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2016] [Revised: 12/30/2016] [Accepted: 02/01/2017] [Indexed: 02/08/2023]
Abstract
BACKGROUND & AIMS Non-alcoholic steatohepatitis (NASH) is a leading cause of chronic liver disease in Western countries. It is unclear how infiltrating leukocytes affect NASH-development. Our study aims to investigate the role of the homing/receptor, pair mucosal addressin cell adhesion molecule-1 (MAdCAM-1)/β7-Integrin, on immune cell recruitment and disease progression in a steatohepatitis model. METHODS Constitutive β7-Integrin deficient (β7-/-) and MAdCAM-1 deficient (MAdCAM-1-/-) mice were fed a high fat diet (HFD) for 26weeks or methionine-choline-deficient-diet (MCD) for 4weeks. RESULTS β7-/- mice displayed earlier and more progressive steatohepatitis during HFD- and MCD-treatment, while MAdCAM-1-/- mice showed less histomorphological changes. The anti-oxidative stress response was significantly weaker in β7-/- mice as reflected by a significant downregulation of the transcription factors nuclear-factor(erythroid-derived 2)-like 2 (Nrf2) and heme-oxigenase-1 (HO-1). Additionally, stronger dihydroethidium-staining revealed an increased oxidative stress response in β7-/- animals. In contrast, MAdCAM-1-/- mice showed an upregulation of the anti-oxidative stress response. β7-/- animals exhibited stronger hepatic infiltration of inflammatory cells, especially neutrophils, reflecting earlier steatohepatitis initiation. Expression of regulatory T cell (TReg) markers as well as numbers of anti-inflammatory macrophages was significantly enhanced in MAdCAM-1-/- mice. Those changes finally resulted in earlier and stronger collagen accumulation in β7-/- mice, whereas MAdCAM-1-/- mice were protected from fibrosis initiation. CONCLUSIONS Adhesion molecule mediated effector cell migration contributes to the outcome of steatohepatitis in the HFD- and the MCD model. While MAdCAM-1 promotes steatohepatitis, β7-Integrin unexpectedly exerts protective effects. β7-/- mice show earlier steatohepatitis initiation and significantly stronger fibrosis progression. Accordingly, the interaction of β7-Integrins and their receptor MAdCAM-1 provide novel targets for therapeutic interventions in steatohepatitis. LAY SUMMARY The mucosal addressin cell adhesion molecule 1 (MAdCAM-1) is expressed in livers upon diet-induced non-alcoholic steatohepatitis (NASH). Loss of MAdCAM-1 has beneficial effects regarding the development of NASH - manifested by reduced hepatic oxidative stress and decreased inflammation. In contrast, β7-Integrin-deficiency results in increased steatohepatitis.
Collapse
Affiliation(s)
- Hannah K Drescher
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Germany
| | - Angela Schippers
- Department of Pediatrics, University Hospital, RWTH Aachen, Germany
| | - Thomas Clahsen
- Department of Pediatrics, University Hospital, RWTH Aachen, Germany
| | - Hacer Sahin
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Germany
| | - Heidi Noels
- Institute of Molecular Cardiovascular Research (IMCAR), University Hospital, RWTH Aachen, Germany
| | - Mathias Hornef
- Institute of Medical Microbiology, University Hospital, RWTH Aachen, Germany
| | - Norbert Wagner
- Department of Pediatrics, University Hospital, RWTH Aachen, Germany
| | - Christian Trautwein
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Germany
| | - Konrad L Streetz
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Germany
| | - Daniela C Kroy
- Department of Internal Medicine III, University Hospital, RWTH Aachen, Germany.
| |
Collapse
|
31
|
Abstract
Various therapeutic advances have led to a paradigm shift in the clinical management of patients with IBD. The introduction of immunosuppressive (such as azathioprine) and biologic agents (such as TNF blockers) has markedly reduced the need to use corticosteroids for therapy. Furthermore, the α4β7 integrin blocker vedolizumab has been introduced for clinical IBD therapy. Moreover, various new inhibitors of cytokines (for example, IL-6-IL-6R and IL-12-IL-23 blockers or apremilast), modulators of cytokine signalling events (for example, JAK inhibitors or SMAD7 blocker), inhibitors of transcription factors (for example, GATA3 or RORγt) and new anti-adhesion and anti-T-cell-activation and migration strategies (for example, β7 integrin, sphingosine 1-phosphate receptors and MAdCAM1 inhibitors, regulatory T-cell therapy and stem cells) are currently being evaluated in controlled clinical trials. This Review aims to provide a comprehensive overview about current and future therapeutic approaches for IBD therapy. Furthermore, potential mechanisms of action of these therapeutic approaches and their implications for clinical therapy in IBD are discussed.
Collapse
Affiliation(s)
- Markus F Neurath
- Department of Medicine 1, Kussmaul Campus for Medical Research, Ludwig Demling Endoscopy Center of Excellence, Ulmenweg 18, University of Erlangen-Nürnberg, 91054 Erlangen, Germany
| |
Collapse
|
32
|
Gidwaney NG, Pawa S, Das KM. Pathogenesis and clinical spectrum of primary sclerosing cholangitis. World J Gastroenterol 2017; 23:2459-2469. [PMID: 28465630 PMCID: PMC5394509 DOI: 10.3748/wjg.v23.i14.2459] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 01/21/2017] [Accepted: 03/20/2017] [Indexed: 02/06/2023] Open
Abstract
Primary sclerosing cholangitis (PSC) is a disease of the biliary tract, which has been documented in the literature since 1867. This disease has a strong predilection for affecting men and can be seen in individuals as young as 2 years of age. PSC has a strong associated with inflammatory bowel disease, more commonly with ulcerative colitis, and is also part of the clinical spectrum of IgG4-related diseases. Small-duct PSC, a variant of PSC, also has an association with inflammatory bowel disease. The exact pathogenesis of PSC is not well understood at present, however, is likely a combination of a genetic predisposition with alteration of the molecular structure of the gut. Abnormal serum liver chemistry and presence of certain autoimmune markers are usually the first indicators leading to a diagnosis of PCS, however, these may often be normal in early stages of this disease. The diagnosis is made by cholangiography, which is now considered the gold standard. PSC is a known pre-malignant condition. Such patients have an increased risk of developing cholangiocarcinoma, gallbladder neoplasia, and colon cancer. Many new treatment modalities have emerged in the recent past, including anti-tumor necrosis factor- α and anti-integrins; however, liver transplantation is the only known cure for PSC. Despite past and present research, PSC remains an enigmatic biliary disease with few viable treatment options.
Collapse
|
33
|
Novel Insights into the Mechanisms of Gut Homing and Antiadhesion Therapies in Inflammatory Bowel Diseases. Inflamm Bowel Dis 2017; 23:617-627. [PMID: 28296823 DOI: 10.1097/mib.0000000000001067] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Therapeutic compounds interfering with T cell trafficking are a new column of inflammatory bowel disease (IBD) treatment. Currently, the anti-α4β7 integrin antibody vedolizumab is successfully used in the clinic and further drugs are likely to follow. Despite these clinical advances, the precise mechanistic background of their action is only gradually elucidated and still a matter of intensive research. Only recently, advances made with the help of new in vivo models and human studies have contributed to shape our concept of T cell trafficking in IBD by deciphering some important and so far unanswered questions. At the same time, basic and clinical data have generated new issues to be addressed on the way toward a clear perception of trafficking mechanisms and toward elucidation of the action of compounds interfering with this process. In this review, we will give a comprehensive outline of all components of T cell trafficking in regard to IBD before discussing the current knowledge concerning targeted interference with integrins in this complex network. Moreover, we will summarize remaining ambiguity and give an outlook on potential future targets.
Collapse
|
34
|
Mason JC, Haskard DO. The Clinical Importance of Leucocyte and Endothelial Cell Adhesion Molecules in Inflammation. ACTA ACUST UNITED AC 2016. [DOI: 10.1177/1358863x9400500306] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
|
35
|
Fischer A, Zundler S, Atreya R, Rath T, Voskens C, Hirschmann S, López-Posadas R, Watson A, Becker C, Schuler G, Neufert C, Atreya I, Neurath MF. Differential effects of α4β7 and GPR15 on homing of effector and regulatory T cells from patients with UC to the inflamed gut in vivo. Gut 2016; 65:1642-1664. [PMID: 26209553 PMCID: PMC5036234 DOI: 10.1136/gutjnl-2015-310022] [Citation(s) in RCA: 124] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2015] [Accepted: 06/22/2015] [Indexed: 12/12/2022]
Abstract
OBJECTIVE Gut homing of lymphocytes via adhesion molecules has recently emerged as new target for therapy in IBDs. We aimed to analyse the in vivo homing of effector (Teff) and regulatory (Treg) T cells to the inflamed gut via α4β7 and G protein receptor GPR15. DESIGN We assessed the expression of homing receptors on T cells in peripheral blood and inflamed mucosa. We studied the migration pattern and homing of Teff and Treg cells to the inflamed gut using intravital confocal microscopy and FACS in a humanised mouse model in dextran sodium sulfate-treated NSG (NOD.Cg-Prkdcscid-Il2rgtm1Wjl/SzJ) mice. RESULTS Expression of GPR15 and α4β7 was significantly increased on Treg rather than Teff cells in peripheral blood of patients with UC as compared with Crohn's disease and controls. In vivo analysis in a humanised mouse model showed augmented gut homing of UC Treg cells as compared with controls. Moreover, suppression of UC (but not control) Teff and Treg cell homing was noted upon treatment with the α4β7 antibody vedolizumab. In contrast, siRNA blockade of GPR15 had only effects on homing of Teff cells but did not affect Treg homing in UC. Clinical vedolizumab treatment was associated with marked expansion of UC Treg cells in peripheral blood. CONCLUSIONS α4β7 rather than GPR15 is crucial for increased colonic homing of UC Treg cells in vivo, while both receptors control UC Teff cell homing. Vedolizumab treatment impairs homing of UC Treg cells leading to their accumulation in peripheral blood with subsequent suppression of systemic Teff cell expansion.
Collapse
Affiliation(s)
- Anika Fischer
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Sebastian Zundler
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Raja Atreya
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Timo Rath
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Caroline Voskens
- Department of Dermatology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Simon Hirschmann
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Rocío López-Posadas
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Alastair Watson
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Christoph Becker
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, University of Erlangen-Nuremberg, Erlangen, Germany
| | - Clemens Neufert
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Imke Atreya
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, University of Erlangen-Nuremberg, Kussmaul Campus for Medical Research & Translational Research Center, Erlangen, Germany
| |
Collapse
|
36
|
Tobi M, Thomas P, Ezekwudo D. Avoiding hepatic metastasis naturally: Lessons from the cotton top tamarin (Saguinus oedipus). World J Gastroenterol 2016; 22:5479-94. [PMID: 27350726 PMCID: PMC4917608 DOI: 10.3748/wjg.v22.i24.5479] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/22/2016] [Revised: 04/26/2016] [Accepted: 05/21/2016] [Indexed: 02/06/2023] Open
Abstract
Much has been written about hepatic metastasis and animal models abound. In terms of the human experience, progress in treating this final common pathway, a terminal event of many human malignancies has been relatively slow. The current thinking is that primary prevention is best served by early detection of cancer and eradication of early stage cancers by screening. Some cancers spread early in their course and the role of screening may be limited. Until relatively recently there has not been a pathfinder model that makes the evasion of this unfortunate event a reality. This review discusses such an animal model and attempts to relate it to human disease in terms of intervention. Concrete proposals are also offered on how scientists may be able to intervene to prevent this deadly progression of the cancer process.
Collapse
|
37
|
Brasseit J, Althaus-Steiner E, Faderl M, Dickgreber N, Saurer L, Genitsch V, Dolowschiak T, Li H, Finke D, Hardt WD, McCoy KD, Macpherson AJ, Corazza N, Noti M, Mueller C. CD4 T cells are required for both development and maintenance of disease in a new mouse model of reversible colitis. Mucosal Immunol 2016; 9:689-701. [PMID: 26376366 DOI: 10.1038/mi.2015.93] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2014] [Accepted: 08/06/2015] [Indexed: 02/04/2023]
Abstract
Current therapies to treat inflammatory bowel diseases have limited efficacy, significant side effects, and often wane over time. Little is known about the cellular and molecular mechanisms operative in the process of mucosal healing from colitis. To study such events, we developed a new model of reversible colitis in which adoptive transfer of CD4(+)CD45RB(hi) T cells into Helicobacter typhlonius-colonized lymphopenic mice resulted in a rapid onset of colonic inflammation that was reversible through depletion of colitogenic T cells. Remission was associated with an improved clinical and histopathological score, reduced immune cell infiltration to the intestinal mucosa, altered intestinal gene expression profiles, regeneration of the colonic mucus layer, and the restoration of epithelial barrier integrity. Notably, colitogenic T cells were not only critical for induction of colitis but also for maintenance of disease. Depletion of colitogenic T cells resulted in a rapid drop in tumor necrosis factor α (TNFα) levels associated with reduced infiltration of inflammatory immune cells to sites of inflammation. Although neutralization of TNFα prevented the onset of colitis, anti-TNFα treatment of mice with established disease failed to resolve colonic inflammation. Collectively, this new model of reversible colitis provides an important research tool to study the dynamics of mucosal healing in chronic intestinal remitting-relapsing disorders.
Collapse
Affiliation(s)
- J Brasseit
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - E Althaus-Steiner
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - M Faderl
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - N Dickgreber
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - L Saurer
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - V Genitsch
- Division of Clinical Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - T Dolowschiak
- Institute of Microbiology, ETH Zürich, Zurich, Switzerland
| | - H Li
- Maurice E. Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - D Finke
- Division of Developmental Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - W-D Hardt
- Institute of Microbiology, ETH Zürich, Zurich, Switzerland
| | - K D McCoy
- Maurice E. Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - A J Macpherson
- Maurice E. Müller Laboratories, University Clinic for Visceral Surgery and Medicine, University of Bern, Bern, Switzerland
| | - N Corazza
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - M Noti
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| | - C Mueller
- Division of Experimental Pathology, Institute of Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
38
|
Sniping the scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases. Pharmacol Res 2016; 107:27-41. [DOI: 10.1016/j.phrs.2016.02.023] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 02/23/2016] [Accepted: 02/23/2016] [Indexed: 02/07/2023]
|
39
|
Petkau JM, Eksteen B. Selective biologics for ulcerative colitis and Crohn's disease - clinical utility of vedolizumab. Biologics 2016; 10:33-52. [PMID: 27022240 PMCID: PMC4790488 DOI: 10.2147/btt.s71679] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Inflammatory bowel disease (IBD) encompasses a cluster of different disease phenotypes which are broadly classified into ulcerative colitis and Crohn’s disease. Disease pathogenesis is driven by abnormal host immune responses to their resident gut microbiome in genetically susceptible individuals. Clinical disease features and outcomes are heterogenous and not unexpected as over 163 genetic loci are associated with disease susceptibility, and there are great variability in environmental exposures. Despite this variability, there has been relatively few efficacious therapies for particularly moderate-to-severe IBD. Treatment has been dominated by antitumor necrosis alpha agents with significant success but equally potentially serious adverse events. Therapeutic targeting of leucocyte trafficking has emerged as a viable alternative therapy, with vedolizumab being the lead compound. This review focuses primarily on its biological function as a selective gut immunotherapy, its safety and efficacy, and its emerging role as a mainstream therapy in managing IBD.
Collapse
Affiliation(s)
- Jill Mv Petkau
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | - Bertus Eksteen
- Snyder Institute for Chronic Diseases, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| |
Collapse
|
40
|
Habtezion A, Nguyen LP, Hadeiba H, Butcher EC. Leukocyte Trafficking to the Small Intestine and Colon. Gastroenterology 2016; 150:340-54. [PMID: 26551552 PMCID: PMC4758453 DOI: 10.1053/j.gastro.2015.10.046] [Citation(s) in RCA: 244] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 10/21/2015] [Accepted: 10/26/2015] [Indexed: 12/14/2022]
Abstract
Leukocyte trafficking to the small and large intestines is tightly controlled to maintain intestinal immune homeostasis, mediate immune responses, and regulate inflammation. A wide array of chemoattractants, chemoattractant receptors, and adhesion molecules expressed by leukocytes, mucosal endothelium, epithelium, and stromal cells controls leukocyte recruitment and microenvironmental localization in intestine and in the gut-associated lymphoid tissues (GALTs). Naive lymphocytes traffic to the gut-draining mesenteric lymph nodes where they undergo antigen-induced activation and priming; these processes determine their memory/effector phenotypes and imprint them with the capacity to migrate via the lymph and blood to the intestines. Mechanisms of T-cell recruitment to GALT and of T cells and plasmablasts to the small intestine are well described. Recent advances include the discovery of an unexpected role for lectin CD22 as a B-cell homing receptor GALT, and identification of the orphan G-protein-coupled receptor 15 (GPR15) as a T-cell chemoattractant/trafficking receptor for the colon. GPR15 decorates distinct subsets of T cells in mice and humans, a difference in species that could affect translation of the results of mouse colitis models to humans. Clinical studies with antibodies to integrin α4β7 and its vascular ligand mucosal vascular addressin cell adhesion molecule 1 are proving the value of lymphocyte trafficking mechanisms as therapeutic targets for inflammatory bowel diseases. In contrast to lymphocytes, cells of the innate immune system express adhesion and chemoattractant receptors that allow them to migrate directly to effector tissue sites during inflammation. We review the mechanisms for innate and adaptive leukocyte localization to the intestinal tract and GALT, and discuss their relevance to human intestinal homeostasis and inflammation.
Collapse
Affiliation(s)
- Aida Habtezion
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California.
| | - Linh P Nguyen
- Division of Gastroenterology and Hepatology, Stanford University School of Medicine, Stanford, California
| | - Husein Hadeiba
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research, Palo Alto, California
| | - Eugene C Butcher
- The Center for Molecular Biology and Medicine, Veterans Affairs Palo Alto Health Care System, The Palo Alto Veterans Institute for Research, Palo Alto, California; Laboratory of Immunology and Vascular Biology, Department of Pathology, Stanford University School of Medicine, Stanford, California.
| |
Collapse
|
41
|
Abstract
Dysregulation of the key genetic, immunologic, and microbiome compounds of the gut-liver axis is the basis for inflammatory bowel disease (IBD) and primary sclerosing cholangitis (PSC). This creates opportunities to accelerate therapies that have been traditionally developed for IBD to be used in PSC to the benefit of both diseases. Shared genetic susceptibility loci has yielded important clues into the pathogenesis of PSC-IBD. Understanding of the critical links between PSC and IBD are essential in designing clinical care pathways for these complex patients.
Collapse
Affiliation(s)
- Bertus Eksteen
- Division of Gastroenterology and Hepatology, Health Research and Innovation Centre (HRIC), Snyder Institute for Chronic Diseases, University of Calgary, 4AC66 - 3280 Hospital Drive Northwest, Calgary, Alberta T2N 4N1, Canada.
| |
Collapse
|
42
|
Vanhove W, Nys K, Vermeire S. Therapeutic innovations in inflammatory bowel diseases. Clin Pharmacol Ther 2015; 99:49-58. [PMID: 26509246 DOI: 10.1002/cpt.286] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2015] [Revised: 10/22/2015] [Accepted: 10/23/2015] [Indexed: 12/19/2022]
Abstract
Inflammatory bowel disease (IBD) is a spectrum of complex multifactorial immune disorders characterized by chronic inflammation of the gut. Significant advances have been made in unraveling the pathogenesis of this disease spectrum, which have spurred the discovery of new therapeutic targets and strategies. In this review, we highlight the emerging new classes of IBD therapeutics under clinical evaluation and their method of action, including JAK inhibitors, anti-SMAD7 oligonucleotides, and cell-based therapies. Moreover, we discuss how an approach based on unique molecular insights in a given patient will, in the future, lead to a truly individualized/tailored disease management, starting at diagnosis, aiding in prognosis, and resulting in a personalized therapeutic approach.
Collapse
Affiliation(s)
- W Vanhove
- Department of Clinical and Experimental Medicine, University Hospitals Leuven, Leuven, Belgium
| | - K Nys
- Department of Clinical and Experimental Medicine, University Hospitals Leuven, Leuven, Belgium
| | - S Vermeire
- Department of Clinical and Experimental Medicine, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
43
|
Mizoguchi A, Takeuchi T, Himuro H, Okada T, Mizoguchi E. Genetically engineered mouse models for studying inflammatory bowel disease. J Pathol 2015; 238:205-19. [PMID: 26387641 DOI: 10.1002/path.4640] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2015] [Revised: 09/05/2015] [Accepted: 09/14/2015] [Indexed: 12/11/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic intestinal inflammatory condition that is mediated by very complex mechanisms controlled by genetic, immune, and environmental factors. More than 74 kinds of genetically engineered mouse strains have been established since 1993 for studying IBD. Although mouse models cannot fully reflect human IBD, they have provided significant contributions for not only understanding the mechanism, but also developing new therapeutic means for IBD. Indeed, 20 kinds of genetically engineered mouse models carry the susceptibility genes identified in human IBD, and the functions of some other IBD susceptibility genes have also been dissected out using mouse models. Cutting-edge technologies such as cell-specific and inducible knockout systems, which were recently employed to mouse IBD models, have further enhanced the ability of investigators to provide important and unexpected rationales for developing new therapeutic strategies for IBD. In this review article, we briefly introduce 74 kinds of genetically engineered mouse models that spontaneously develop intestinal inflammation.
Collapse
Affiliation(s)
- Atsushi Mizoguchi
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Takahito Takeuchi
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Hidetomo Himuro
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Toshiyuki Okada
- Department of Immunology, Kurume University School of Medicine, 67 Asahi-machi, Kurume, Fukuoka, 830-0011, Japan
| | - Emiko Mizoguchi
- Gastrointestinal Unit and Center for the Study of Inflammatory Bowel Disease, Massachusetts General Hospital, Harvard Medical School, Fruit Street, Boston, MA, 02114, USA
| |
Collapse
|
44
|
Hagan M, Cross RK. Safety of vedolizumab in the treatment of Crohn's disease and ulcerative colitis. Expert Opin Drug Saf 2015; 14:1473-9. [PMID: 26138111 DOI: 10.1517/14740338.2015.1063612] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
INTRODUCTION Vedolizumab is the latest FDA-approved anti-integrin therapy for treatment of moderate-to-severe inflammatory bowel disease (IBD). The safety and efficacy of vedolizumab have been studied in short-term clinical trials. AREAS COVERED This paper reviews the safety profile of vedolizumab compared with other biologics. It also highlights the mechanism of action of the medication. We discuss the current position of vedolizumab in our current algorithm for IBD management and comment on future prospects of the drug. EXPERT OPINION Vedolizumab appears to be a safe and effective option in the treatment of moderate-to-severe IBD in the short term. Long-term observational studies and post-marketing safety data are needed to ascertain the long-term efficacy and side effect profile.
Collapse
Affiliation(s)
- Matilda Hagan
- a 1 University of Maryland, Division of Gastroenterology and Hepatology, Department of Medicine , 100 North Greene Street, Lower Level, Baltimore, MD 21201, USA
| | | |
Collapse
|
45
|
Crohn’s Disease. Mucosal Immunol 2015. [DOI: 10.1016/b978-0-12-415847-4.00082-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
46
|
Arseneau KO, Cominelli F. Targeting leukocyte trafficking for the treatment of inflammatory bowel disease. Clin Pharmacol Ther 2014; 97:22-8. [PMID: 25670380 DOI: 10.1002/cpt.6] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2014] [Accepted: 10/10/2014] [Indexed: 12/17/2022]
Abstract
Inflammatory bowel disease (IBD) is a chronic immune-mediated inflammatory disease of the intestine that includes both Crohn's disease and ulcerative colitis, and afflicts nearly 1 million people throughout North America. As our understanding of IBD pathogenesis grows, several new therapies have been developed that use monoclonal antibodies to specifically target key mediators and biological pathways implicated in IBD immune dysfunction. One important pathway involves leukocyte trafficking and infiltration into the affected intestinal tissues. This review provides a summary of the different therapies that have been developed to inhibit leukocyte trafficking to the inflamed gut, and evaluates the relative safety and efficacy of these novel drugs within the context of existing medical therapies for IBD.
Collapse
Affiliation(s)
- K O Arseneau
- Department of Medicine, Case Western Reserve University, Cleveland, Ohio, USA and Digestive Health Research Center, Case Western Reserve University, Cleveland, Ohio, USA
| | | |
Collapse
|
47
|
Kurmaeva E, Lord JD, Zhang S, Bao JR, Kevil CG, Grisham MB, Ostanin DV. T cell-associated α4β7 but not α4β1 integrin is required for the induction and perpetuation of chronic colitis. Mucosal Immunol 2014; 7:1354-65. [PMID: 24717354 PMCID: PMC4417258 DOI: 10.1038/mi.2014.22] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2013] [Revised: 02/17/2014] [Accepted: 03/04/2014] [Indexed: 02/04/2023]
Abstract
Anti-adhesion therapies that target α(4) integrins (e.g., natalizumab) are thought to work by blocking T-cell recruitment to the intestinal tissues in patients with Crohn's disease (CD); however, little direct evidence is available to confirm this contention. We wished to evaluate the importance of T cell-associated α(4) integrins in a chronic colitis model in mice and to determine the effect of natalizumab treatment on intestinal tissue T-cell accumulation in human CD. Adoptive transfer of T cells lacking α(4) (α(4)(-/-)) but not β(1) integrin into immunodeficient mice produced significantly attenuated disease. This was correlated with reduced numbers of colon CD4 T cells compared with the control mice; however, tissue distribution of T helper type 1 (Th1) and T helper type 17 (Th17) cells and regulatory T cells (Tregs) was not affected by the lack of α(4). Furthermore, α(4)(-/-) T cells demonstrated defective homing to the chronically inflamed small intestines and colons. Finally, patients treated with natalizumab showed significant reduction in mucosal CD4 T cells and no skewing in the foxp3(+) Treg or T-bet(+)Th1 fractions thereof. These results demonstrate a direct role for T cell-associated α(4)β(7) but not α(4)β(1) integrins during initiation and perpetuation of chronic colitis. Moreover, our data demonstrated that natalizumab treatment reduced mucosal CD4 T-cell accumulation in CD patients.
Collapse
Affiliation(s)
- E Kurmaeva
- Center of Excellence for Arthritis and Rheumatology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - JD Lord
- Translational Research Program, Benaroya Research Institute, Seattle, Washington, USA
| | - S Zhang
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - JR Bao
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - CG Kevil
- Department of Pathology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| | - MB Grisham
- Department of Immunology and Molecular Microbiology, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - DV Ostanin
- Center of Excellence for Arthritis and Rheumatology, Louisiana State University Health Sciences Center, Shreveport, Louisiana, USA
| |
Collapse
|
48
|
Beniwal-Patel P, Saha S. The role of integrin antagonists in the treatment of inflammatory bowel disease. Expert Opin Biol Ther 2014; 14:1815-23. [PMID: 25288228 DOI: 10.1517/14712598.2014.967209] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Ulcerative colitis (UC) and Crohn's disease are chronic inflammatory diseases of the bowel associated with complex inflammatory cascades within the mucosal lining of the gut. AREAS COVERED INTEGRINS and their use as therapies in UC and Crohn's. EXPERT OPINION The anti-adhesion molecules are a welcome addition to the armamentarium of medical therapies for inflammatory bowel disease.
Collapse
Affiliation(s)
- Poonam Beniwal-Patel
- University of Wisconsin School of Medicine and Public Health , 1685 Highland Avenue Madison, WI 53705 , USA
| | | |
Collapse
|
49
|
Abstract
: The etiologies of Crohn's disease and ulcerative colitis, the 2 major forms of inflammatory bowel disease in humans, remain unknown, but experimental studies suggest that inflammatory bowel disease results from interaction between environmental and genetic factors, which promotes an exaggerated and inappropriately controlled inflammatory response that is directed against normal components of the gut flora. There is also evidence that tissue damage is due to a dynamic interplay between immune and nonimmune cells, and recruitment of lymphocytes from the blood stream to the gut wall is crucial for amplifying and sustaining the ongoing mucosal inflammation. These advances have led to the development of several compounds blocking gut homing of effector lymphocytes, which have recently been used or are now ready to move into clinical practice. This article summarizes the recent data on the use of integrin-targeting and adhesion molecule-targeting therapeutics to attenuate the detrimental inflammatory response in inflammatory bowel disease.
Collapse
|
50
|
Halpert G, Sredni B. The effect of the novel tellurium compound AS101 on autoimmune diseases. Autoimmun Rev 2014; 13:1230-5. [PMID: 25153485 DOI: 10.1016/j.autrev.2014.08.003] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2014] [Accepted: 07/30/2014] [Indexed: 02/06/2023]
Abstract
Tellurium is a rare element, which has been regarded as a non-essential trace element despite its relative abundance in the human body. The chemistry of tellurium supports a plethora of activities, but its biochemistry is not clearly established to date. The small tellurium(IV) compound, ammonium trichloro (dioxoethylene-o,o')tellurate (AS101) developed and initially investigated by us, is currently being evaluated in Phase II clinical trials in psoriasis patients. AS101 is the first tellurium compound to be tested for clinical efficacy. This compound is a potent immunomodulator both in vitro and in vivo with a variety of potential therapeutic applications. The present review will focus on the immunomodulatory properties of AS101, and specifically, its effects in mitigating autoimmune diseases. AS101 has several activities that act on the immune system, including: 1) its ability to reduce IL-17 levels and to inhibit the function of Th17 cells; 2) its specific unique redox-modulating activities enabling the inhibition of specific leukocyte integrins such as α4β1 and α4β7, that are pivotal for diapedesis of macrophages and CD4(+) T inflammatory/auto-reactive cells into the autoimmune tissues; and 3) its ability to enhance the activity of regulatory T cells (Treg). These activities coupled with its excellent safety profile suggest that AS101 may be a promising candidate for the management of autoimmune diseases.
Collapse
Affiliation(s)
- Gilad Halpert
- C.A.I.R. Institute, The Safdié AIDS and Immunology Research Center, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel
| | - Benjamin Sredni
- C.A.I.R. Institute, The Safdié AIDS and Immunology Research Center, The Mina & Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan 52900, Israel.
| |
Collapse
|