1
|
Bernardo L, Solana JC, Sánchez C, Torres A, Reyes-Cruz EY, Carrillo E, Moreno J. Immunosuppressants alter the immune response associated with Glucantime ® treatment for Leishmania infantum infection in a mouse model. Front Immunol 2023; 14:1285943. [PMID: 38106411 PMCID: PMC10722182 DOI: 10.3389/fimmu.2023.1285943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/16/2023] [Indexed: 12/19/2023] Open
Abstract
Background Immunosuppression is a major risk factor for the development of visceral leishmaniasis (VL). The number of patients receiving immunosuppressant drugs such as TNF antagonist (anti-TNF) and methotrexate (MTX) is increasing. In these patients, VL is more severe, their response to treatment poorer, and they are at higher risk of relapse, a consequence (largely) of the poor and inappropriate immune response they develop. Objectives To examine the effect of immunosuppressive treatment on the host immune response and thus gain insight into the reduced efficacy of pentavalent antimonials in these patients. Experiments were performed using BALB/c mice immunosuppressed with anti-TNF or MTX, infected with Leishmania infantum promastigotes, and then treated with Glucantime® at clinical doses. Results Immunosuppression with both agents impeded parasite elimination from the spleen and bone marrow. Low pro-inflammatory cytokine production by CD4+ and CD8+ T cells was detected, along with an increase in PD-1 and IL-10 expression by B and T cells in the immunosuppressed groups after treatment. Conclusion The immunosuppressed mice were unable to develop specific cellular immunity to the parasite, perhaps explaining the greater risk of VL relapse seen in pharmacologically immunosuppressed human patients.
Collapse
Affiliation(s)
- Lorena Bernardo
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Jose Carlos Solana
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Carmen Sánchez
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
| | - Ana Torres
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Eder Yaveth Reyes-Cruz
- LADISER Immunology and Molecular Biology, Faculty of Chemical Sciences, Universidad Veracruzana, Orizaba, Veracruz, Mexico
| | - Eugenia Carrillo
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| | - Javier Moreno
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
2
|
Cannon L, Pan A, Kovalick L, Sarkissian A, Wu EY. Secondary immunodeficiencies and infectious considerations of biologic immunomodulatory therapies. Ann Allergy Asthma Immunol 2023; 130:718-726. [PMID: 36801438 PMCID: PMC10247415 DOI: 10.1016/j.anai.2023.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/02/2023] [Accepted: 02/13/2023] [Indexed: 02/19/2023]
Abstract
Biologic immunomodulatory medications have rapidly expanded in the previous decades, providing new treatment options for individuals with a spectrum of oncologic, allergic, rheumatologic, and neurologic conditions. Biologic therapies alter immune function and can impair key host defense mechanisms, resulting in secondary immunodeficiency and increased infectious risks. Biologic medications can increase general risk for upper respiratory tract infections but can also be associated with unique infectious risks owing to distinct mechanisms of action. With the widespread use of these medications, providers in every area of medicine will likely care for individuals receiving biologic therapies and understanding their potential infectious complications can help mitigate these risks. This practical review discusses the infectious implications of biologics by class of medication and provides recommendations regarding the examination and screening both before therapy initiation and while the patient is receiving the medication. With this knowledge and background, providers can reduce risk whereas patients receive the treatment benefits of these biologic medications.
Collapse
Affiliation(s)
- Laura Cannon
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alice Pan
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Department of Pharmacy, UNC Health, Chapel Hill, North Carolina
| | - Leonard Kovalick
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Aliese Sarkissian
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Eveline Y Wu
- Division of Pediatric Rheumatology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina; Division of Pediatric Allergy/Immunology, Department of Pediatrics, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina.
| |
Collapse
|
3
|
Chung J, Choi MR, Kim MG, Park SK, Kim YM. Abatacept (Cytotoxic T Lymphocyte Antigen 4-Fragment Crystallizable) Reduces Allergic Inflammation of Ovalbumin-Sensitized Mice. Am J Rhinol Allergy 2022; 36:432-439. [PMID: 34994213 DOI: 10.1177/19458924211072364] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/08/2023]
Abstract
BACKGROUND Abatacept (Aba) is a cytotoxic T-lymphocyte antigen-4 and fragment crystallizable fusion protein. Aba blocks B7/Cluster of differentiation 28 - cytotoxic T-lymphocyte antigen-4 costimulatory pathway, inhibits cluster of differentiation 4+ T-cell activation, and is used as an anti-inflammatory drug. OBJECTIVES We conducted this study to assess the effectiveness of Aba in the treatment of allergic rhinitis (AR) in a mouse model. METHODS We divided 40 four-week-old BALB/c mice into four groups: control group (n = 10), positive control group (AR, n = 10), Aba group (AR + Aba, n = 10), and dexamethasone group (AR + Dex, n = 10). Mice in each group were challenged intranasally with daily ovalbumin (OVA) administration. Episodes of sneezing and nose rubbing were counted. Mice were sacrificed on day 42 and cytokines were measured in nasal lavage fluid. Nasal mucosae of five mice from each group were used for reverse transcriptase-polymerase chain reaction and western blot assay. Samples were collected from five mice from each group for histological analysis. RESULTS Symptoms of AR significantly improved in the AR + Aba and AR + Dex groups compared with the AR group. Fewer eosinophils and goblet cells were seen in the AR + Aba and AR + Dex groups compared with the AR group. Both the AR + Aba and AR + Dex groups showed a significant decrease in nasal T helper 2 cytokine levels, including interleukin (IL)-4, IL-5, IL-13 and T cell activation related IL-17A, and interferon gamma (IFN- γ). Total immunoglobulin (Ig) E and OVA-specific IgG1 levels were also significantly lower in the AR + Aba and AR + Dex groups. OVA-specific IgE level was also significantly lower in the AR + Aba than AR group. CONCLUSIONS Aba suppresses allergic inflammation and appears to be a good treatment for AR.
Collapse
Affiliation(s)
- Jaein Chung
- Chungnam National University School of Medicine, Daejeon, Korea
| | - Mi-Ra Choi
- Chungnam National University School of Medicine, Daejeon, Korea
| | - Min Gyu Kim
- Chungnam National University School of Medicine, Daejeon, Korea
| | - Soo Kyoung Park
- Chungnam National University School of Medicine, Daejeon, Korea
| | - Yong Min Kim
- Chungnam National University School of Medicine, Daejeon, Korea
| |
Collapse
|
4
|
Banno S, Yamaguchi E, Iwagaitsu S, Nobata H, Yamaguchi M, Sugiyama H, Kinashi H, Katsuno T, Kubo A, Ito S, Ito Y. Long-term good outcome of the fibrocavitary form of pulmonary Mycobacterium avium complex disease with concomitant abatacept monotherapy in a patient with rheumatoid arthritis. Mod Rheumatol Case Rep 2022; 6:1-5. [PMID: 34637523 DOI: 10.1093/mrcr/rxab002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 03/29/2021] [Accepted: 05/10/2021] [Indexed: 06/13/2023]
Abstract
A 53-year-old woman diagnosed with rheumatoid arthritis (RA) demonstrated thick-walled large cavities with consolidation in the left upper lobe on chest computed tomography (CT). Mycobacterium avium was isolated from sputum cultures, and she was diagnosed as having the fibrocavitary (FC) form of pulmonary Mycobacterium avium complex (MAC) disease. Clarithromycin-containing, multidrug, anti-MAC chemotherapy was started immediately. After 7 months, the cavitary lesions improved, and sputum cultures showed negative conversion. Thereafter, abatacept monotherapy was started due to high RA disease activity. Clinical remission of RA has been sustained and cavitary lesions disappeared by concomitant abatacept and anti-MAC therapy for more than 5 years. Immediate initiation of anti-MAC therapy and prior confirmed efficacy are needed for the treatment of the FC form. Abatacept and anti-MAC therapy could be continued, leading to the withdrawal of prednisolone, along with careful observation by strict chest CT evaluation and repeated sputum cultures. Biologics are generally contraindicated for pulmonary MAC disease, particularly the FC form. When there is a pre-existing lung lesion apparently of FC type, abatacept cannot be started without prior anti-MAC chemotherapy. This case suggests that abatacept may be carefully used to avoid progressive joint destruction after FC lesions of pulmonary MAC disease are resolved.
Collapse
Affiliation(s)
- Shogo Banno
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Etsuro Yamaguchi
- Department of Respiratory Medicine and Allergology, Aichi Medical University, Nagakute, Japan
| | - Shiho Iwagaitsu
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Hironobu Nobata
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Makoto Yamaguchi
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Hirokazu Sugiyama
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Hiroshi Kinashi
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Takayuki Katsuno
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| | - Akihito Kubo
- Department of Respiratory Medicine and Allergology, Aichi Medical University, Nagakute, Japan
| | - Satoru Ito
- Department of Respiratory Medicine and Allergology, Aichi Medical University, Nagakute, Japan
| | - Yasuhiko Ito
- Department of Rheumatology and Nephrology, Aichi Medical University, Nagakute, Japan
| |
Collapse
|
5
|
Higashiguchi M, Yamazaki F, Nishioka K, Kimura H, Matsumoto T. Abatacept for rheumatoid arthritis complicated by disseminated cryptococcosis: a case report. Mod Rheumatol Case Rep 2021; 5:229-235. [PMID: 33843457 DOI: 10.1080/24725625.2021.1909214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
A 78-year-old man developed disseminated cryptococcosis with central nervous system involvement as encapsulated yeast cells were detected in transbronchial biopsy and skin biopsy specimens, and cerebrospinal fluid. Cryptococcus neoformans was confirmed by culture. He had been treated with low-dose prednisolone and methotrexate for rheumatoid arthritis. He started receiving antifungal therapy with intravenous liposomal amphotericin B followed by oral fluconazole. Methotrexate was discontinued. Approximately 4 months after the course of intravenous liposomal amphotericin B was completed, he complained of pain and swelling of the right wrist, which suggested that rheumatoid arthritis was worsening. Abatacept therapy was initiated along with antifungal therapy, and his symptoms relieved. After 24 months of antifungal therapy, although he was still receiving oral fluconazole, he was doing well and the serum cryptococcal antigen had become negative. Disseminated cryptococcosis is an important opportunistic infection associated with low-dose methotrexate for rheumatoid arthritis. Abatacept therapy may be feasible in strictly selected patients with rheumatoid arthritis complicated with cryptococcosis concomitantly with intensive anti-fungal therapy.
Collapse
Affiliation(s)
- Masayoshi Higashiguchi
- Department of Internal Medicine, Osaka Anti-Tuberculosis Association Osaka Hospital, Neyagawa, Osaka, Japan
| | - Fumikazu Yamazaki
- Department of Dermatology, Kansai Medical University, Hirakata, Osaka, Japan
| | - Koji Nishioka
- Department of Internal Medicine, Osaka Anti-Tuberculosis Association Osaka Hospital, Neyagawa, Osaka, Japan
| | - Hiromi Kimura
- Department of Internal Medicine, Osaka Anti-Tuberculosis Association Osaka Hospital, Neyagawa, Osaka, Japan
| | - Tomoshige Matsumoto
- Department of Internal Medicine, Osaka Anti-Tuberculosis Association Osaka Hospital, Neyagawa, Osaka, Japan
| |
Collapse
|
6
|
Bernardo L, Solana JC, Romero-Kauss A, Sánchez C, Carrillo E, Moreno J. Effect of immunosuppressants on the parasite load developed in, and immune response to, visceral leishmaniasis: A comparative study in a mouse model. PLoS Negl Trop Dis 2021; 15:e0009126. [PMID: 33524030 PMCID: PMC7877784 DOI: 10.1371/journal.pntd.0009126] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/11/2021] [Accepted: 01/12/2021] [Indexed: 11/29/2022] Open
Abstract
The increasing use of immunosuppressants in areas where visceral leishmaniasis (VL) is endemic has increased the number of people susceptible to developing more severe forms of the disease. Few studies have examined the quality of the immune response in immunosuppressed patients or experimental animals with VL. The present work characterises the parasite load developed in, and immune response to, Leishmania infantum-induced VL in C57BL/6 mice that, prior to and during infection, received immunosuppressant treatment with methylprednisolone (MPDN), anti-tumour necrosis factor (anti-TNF) antibodies, or methotrexate (MTX). The latter two treatments induced a significant reduction in the number of CD4+ T lymphocytes over the infection period. The anti-TNF treatment was also associated with a higher parasite load in the liver and a lower parasite load in the spleen. This, plus a possibly treatment-induced reduction in the number of cytokine-producing Th1 cells in the spleen, indicates the development of more severe VL. Interestingly, the MPDN and (especially) MTX treatments provoked a greater presence of soluble Leishmania antigen-specific multi-cytokine-producing T cells in the spleen and a lower liver parasite load than in control animals. These results highlight the need to better understand how immunosuppressant treatments might influence the severity of VL in human patients.
Collapse
Affiliation(s)
- Lorena Bernardo
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda (Madrid), Spain
| | - Jose Carlos Solana
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda (Madrid), Spain
| | - Alba Romero-Kauss
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda (Madrid), Spain
| | - Carmen Sánchez
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda (Madrid), Spain
| | - Eugenia Carrillo
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda (Madrid), Spain
| | - Javier Moreno
- WHO Collaborating Centre for Leishmaniasis, National Center for Microbiology, Instituto de Salud Carlos III, Majadahonda (Madrid), Spain
| |
Collapse
|
7
|
Evangelatos G, Koulouri V, Iliopoulos A, Fragoulis GE. Tuberculosis and targeted synthetic or biologic DMARDs, beyond tumor necrosis factor inhibitors. Ther Adv Musculoskelet Dis 2020; 12:1759720X20930116. [PMID: 32612710 PMCID: PMC7309385 DOI: 10.1177/1759720x20930116] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 05/07/2020] [Indexed: 12/12/2022] Open
Abstract
Patients with autoimmune rheumatic diseases (ARD) have an increased risk for tuberculosis (TB). The use of tumor necrosis factor inhibitors (TNFi) and glucocorticoids in these patients has been associated with an increased prevalence of latent TB reactivation. Over the last few years, several biologic disease-modifying anti-rheumatic drugs (bDMARDs), other than TNFi (e.g. rituximab, abatacept, tocilizumab, secukinumab) and targeted synthetic DMARDs (tsDMARDs) [e.g. apremilast, Janus kinase (JAK) inhibitors] have been used for the treatment of patients with ARD. For many of these drugs, especially the newer ones like JAK inhibitors or antibodies against interleukin (IL)-23, most data stem from randomized clinical trials and few are available from real life clinical experience. We sought to review the current evidence for TB risk in patients with ARD treated with tsDMARDs or bDMARDs, other than TNFi. It seems that some of these drugs are associated with a lower TB risk, indirectly compared with TNFi treatment. In fact, it appears that rituximab, apremilast and inhibitors of IL-17 and IL-23 might be safer, while more data are needed for JAK inhibitors. As seen in TNFi, risk for TB is more pronounced in TB-endemic areas. Screening for latent TB must precede initiation of any tsDMARDs or bDMARDs. The growing use of non-TNFi agents has raised the need for more real-life studies that would compare the risk for TB between TNFi and other treatment modalities for ARD. Knowledge about the TB-safety profile of these drugs could help in the decision of drug choice in patients with confirmed latent TB infection or in TB endemic areas.
Collapse
Affiliation(s)
- Gerasimos Evangelatos
- Rheumatology Department, 417 Army Share Fund Hospital (NIMTS), Monis Petraki 10-12, Athens, 11521, Greece
| | - Vasiliki Koulouri
- Department of Physiology, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Alexios Iliopoulos
- Rheumatology Department, 417 Army Share Fund Hospital (NIMTS), Athens, Greece
| | - George E Fragoulis
- Rheumatology Unit, First Department of Propaedeutic Internal Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Greece
- Institute of Infection, Immunity and Inflammation, University of Glasgow, Glasgow, UK
| |
Collapse
|
8
|
Davis JS, Ferreira D, Paige E, Gedye C, Boyle M. Infectious Complications of Biological and Small Molecule Targeted Immunomodulatory Therapies. Clin Microbiol Rev 2020; 33:e00035-19. [PMID: 32522746 PMCID: PMC7289788 DOI: 10.1128/cmr.00035-19] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The past 2 decades have seen a revolution in our approach to therapeutic immunosuppression. We have moved from relying on broadly active traditional medications, such as prednisolone or methotrexate, toward more specific agents that often target a single receptor, cytokine, or cell type, using monoclonal antibodies, fusion proteins, or targeted small molecules. This change has transformed the treatment of many conditions, including rheumatoid arthritis, cancers, asthma, and inflammatory bowel disease, but along with the benefits have come risks. Contrary to the hope that these more specific agents would have minimal and predictable infectious sequelae, infectious complications have emerged as a major stumbling block for many of these agents. Furthermore, the growing number and complexity of available biologic agents makes it difficult for clinicians to maintain current knowledge, and most review articles focus on a particular target disease or class of agent. In this article, we review the current state of knowledge about infectious complications of biologic and small molecule immunomodulatory agents, aiming to create a single resource relevant to a broad range of clinicians and researchers. For each of 19 classes of agent, we discuss the mechanism of action, the risk and types of infectious complications, and recommendations for prevention of infection.
Collapse
Affiliation(s)
- Joshua S Davis
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- Global and Tropical Health Division, Menzies School of Health Research and Charles Darwin University, Darwin, NT, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| | - David Ferreira
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
| | - Emma Paige
- Department of Infectious Diseases, Alfred Hospital, Melbourne, VIC, Australia
| | - Craig Gedye
- School of Medicine, University of New South Wales, Sydney, NSW, Australia
- Department of Oncology, Calvary Mater Hospital, Newcastle, NSW, Australia
| | - Michael Boyle
- Department of Infectious Diseases and Immunology, John Hunter Hospital, Newcastle, NSW, Australia
- School of Medicine and Public Health, University of Newcastle, Newcastle, NSW, Australia
| |
Collapse
|
9
|
Mesquita KC, Dantas TS, de Barros Silva PG, de Queiroz Rodrigues MI, Alves APNN, Mota MRL, Sousa FB. Abatacept treatment impairs the cell migration and wound healing of oral ulcers in rats: Role of interleukin (IL)-1β, -6 and -10 and CD8/CD30 cells: Influence of abatacept treatment on oral wound healing: Experimental model on rats. Life Sci 2020; 243:117243. [PMID: 31917994 DOI: 10.1016/j.lfs.2019.117243] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 12/19/2019] [Accepted: 12/27/2019] [Indexed: 12/01/2022]
Abstract
The present study aimed to evaluate the healing process of ulcers in the jugal mucosa of Wistar rats treated with abatacept. The rats were randomly assigned to four groups: saline-treated control (0.3 mL/kg) abatacept-treated groups at dosages of 3.2, 8.0 and 20.0 mg/kg/week. After two weeks of subcutaneous (SC) administration, ulcers were introduced into the left jugal mucosa with an 8-mm diameter punch. SC administration was continued until euthanasia (after 1, 3, 7, 14 and 21 days of ulceration), and ulcers were clinically measured and animals weighed. Histological slides were evaluated (healing scores and polymorphonuclear, mononuclear, vessel, and fibroblast/myofibroblast counts). We also performed collagenesis analysis (Picrosirius Red) and immunohistochemistry (induced nitric oxide synthase (iNOS), interleukin (IL)-1beta (1β), -6, -10, plus the analysis of CD8 and CD30). The experiment was repeated to perform a vascular permeability assay. ANOVA 1-way or 2-way/Bonferroni and Kruskal-Wallis/Dunn tests were used for statistical analysis (GraphPad Prism 5.0®, p < 0.05). Abatacept treatment reduced the ulcer diameter and the numbers of polymorphonuclear and mononuclear cells; reduced the CD8+/CD30+ ratio and vascular permeability; and increased collagenesis and IL-10 expression at the beginning of the protocol. At the highest dose, there was a delay in repair and vascular proliferation; a reduction in the number of fibroblasts/myofibroblasts; and prolongation of iNOS, IL- and IL- expression. We conclude that abatacept accelerates the healing of oral ulcers by reducing the migration of inflammatory cells, but overdose of abatacept leads to delayed repair and prolongation of proinflammatory cytokine expression.
Collapse
Affiliation(s)
- Karine Cestaro Mesquita
- Division of Oral Pathology, Federal University of Ceará, Fortaleza, Ceará, Brazil; Department of Dentistry, Unichristus, Fortaleza, Ceará, Brazil
| | - Thinali Sousa Dantas
- Division of Oral Pathology, Federal University of Ceará, Fortaleza, Ceará, Brazil; Department of Dentistry, Unichristus, Fortaleza, Ceará, Brazil
| | - Paulo Goberlânio de Barros Silva
- Division of Oral Pathology, Federal University of Ceará, Fortaleza, Ceará, Brazil; Department of Dentistry, Unichristus, Fortaleza, Ceará, Brazil
| | | | | | | | - Fabrício Bitu Sousa
- Division of Oral Pathology, Federal University of Ceará, Fortaleza, Ceará, Brazil; Department of Dentistry, Unichristus, Fortaleza, Ceará, Brazil
| |
Collapse
|
10
|
Diener C, Horneff G. Comparison of adverse events of biologicals for treatment of juvenile idiopathic arthritis: a systematic review. Expert Opin Drug Saf 2019; 18:719-732. [PMID: 31204508 DOI: 10.1080/14740338.2019.1632288] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Objectives: Treatment of juvenile idiopathic arthritis has changed rapidly since the introduction of various biologics almost twenty years ago. Many clinical trials have been performed to monitor efficacy and safety of new agents. The aim of this review is to summarize safety concerns, which were observed during prospective clinical trials. Methods: Since etanercept was the first biologic approved and remains the most frequently used, as first biologic in polyarticular JIA patients, the authors calculated the relative risk of the adverse events for all examined biologicals and compared the values with the value of etanercept. Results: Relative rates for all adverse events showed similar rates for etanercept, infliximab, golimumab, and tocilizumab, whereas adalimumab showed higher rates and abatacept lower rates. Comparison of rates for serious adverse events demonstrated, that rates seemed comparable for etanercept, adalimumab, infliximab, and tocilizumab. Again, abatacept showed a lower rate, whereas golimumab seems to have a higher relative risk for serious adverse events. Rate of infection was lowest in patients treated with abatacept or tocilizumab, patients treated with etanercept, adalimumab and Infliximab again had similar rates. Conclusion: The safety profiles of actually approved biologics are highly acceptable. However, further observation, especially long-term observation through registry studies, is required.
Collapse
Affiliation(s)
- C Diener
- a Department of General Paediatrics, Asklepios Klinik Sankt Augustin , Sankt Augustin , Germany
| | - G Horneff
- a Department of General Paediatrics, Asklepios Klinik Sankt Augustin , Sankt Augustin , Germany.,b Department of Paediatric and Adolescents Medicine, Medical Faculty, University Hospital of Cologne , Cologne , Germany
| |
Collapse
|
11
|
The YIN and YANG of lipoproteins in developing and preventing infectious arthritis by Staphylococcus aureus. PLoS Pathog 2019; 15:e1007877. [PMID: 31226163 PMCID: PMC6608979 DOI: 10.1371/journal.ppat.1007877] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Revised: 07/03/2019] [Accepted: 05/28/2019] [Indexed: 12/21/2022] Open
Abstract
Rapid bone destruction often leads to permanent joint dysfunction in patients with septic arthritis, which is mainly caused by Staphylococcus aureus (S. aureus). Staphylococcal cell wall components are known to induce joint inflammation and bone destruction. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis through TLR2. Arthritis was characterized by rapid infiltration of neutrophils and monocytes. The arthritogenic effect was mediated mainly by macrophages/monocytes and partially via TNF-α but not by neutrophils. Surprisingly, a S. aureus mutant lacking Lpp diacylglyceryl transferase (lgt) caused more severe joint inflammation, which coincided with higher bacterial loads of the lgt mutant in local joints than those of its parental strain. Coinjection of pathogenic S. aureus LS-1 with staphylococcal Lpps into mouse knee joints caused improved bacterial elimination and diminished bone erosion. The protective effect of the Lpps was mediated by their lipid moiety and was fully dependent on TLR2 and neutrophils. The blocking of CXCR2 on neutrophils resulted in total abrogation of the protective effect of the Lpps. Our data demonstrate that S. aureus Lpps elicit innate immune responses, resulting in a double-edged effect. On the one hand, staphylococcal Lpps boost septic arthritis. On the other hand, Lpps act as adjuvants and activate innate immunity, which could be useful for combating infections with multiple drug-resistant strains. Rapid bone destruction often leads to permanent joint dysfunction in septic arthritis, which is mainly caused by S. aureus. Despite advances in the use of antibiotics, permanent reductions in joint function occur in up to 50% of patients, who may also need joint replacement surgery. Additional challenge is posed by increasing antibiotic resistance of S. aureus, causing significant clinical and economic consequences. Although the outcome is poor, the current treatments for septic arthritis remain unchanged since many decades. It is largely unknown which bacterial factors cause aggressive joint damage. Here, we show that a single intra-articular injection of S. aureus lipoproteins (Lpps) into mouse knee joints induced chronic destructive macroscopic arthritis, and the monocytes/macrophages were the main culprit. However, coinjection of pathogenic S. aureus with Lpps into mouse knee joints attenuated the disease. The protective effect of Lpps was mediated by their lipid moiety, TLR2 on the host cells, neutrophil chemokine release, and consequent neutrophil recruitment. Our finding might be used as a novel concept in the treatment of multidrug-resistant bacterial infections.
Collapse
|
12
|
Henkle E, Winthrop KL. Immune Dysfunction and Nontuberculous Mycobacterial Disease. NONTUBERCULOUS MYCOBACTERIAL DISEASE 2019. [DOI: 10.1007/978-3-319-93473-0_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
13
|
Concomitant use of intravenous methylprednisolone to increase retention rate of abatacept in rheumatoid arthritis. Rheumatol Int 2018; 38:1825-1831. [DOI: 10.1007/s00296-018-4115-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Accepted: 07/24/2018] [Indexed: 11/27/2022]
|
14
|
Jeong SH, Nam Y, Jung H, Kim J, Rim YA, Park N, Lee K, Choi S, Jang Y, Kim Y, Moon JH, Jung SM, Park SH, Ju JH. Interrupting oral infection of Porphyromonas gingivalis with anti-FimA antibody attenuates bacterial dissemination to the arthritic joint and improves experimental arthritis. Exp Mol Med 2018; 50:e460. [PMID: 29568073 PMCID: PMC5898898 DOI: 10.1038/emm.2017.301] [Citation(s) in RCA: 25] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 08/29/2017] [Accepted: 09/26/2017] [Indexed: 12/29/2022] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease that typically results in strong inflammation and bone destruction in the joints. It is generally known that the pathogenesis of RA is linked to cardiovascular and periodontal diseases. Though rheumatoid arthritis and periodontitis share many pathologic features such as a perpetual inflammation and bone destruction, the precise mechanism underlying a link between these two diseases has not been fully elucidated. Collagen-induced arthritis (CIA) mice were orally infected with Porphyromonas gingivalis (Pg) or Pg preincubated with an anti-FimA antibody (FimA Ab) specific for fimbriae that are flexible appendages on the cell surface. Pg-infected CIA mice showed oral microbiota disruption and increased alveolar bone loss and had synovitis and joint bone destruction. However, preincubation with FimA Ab led to a significant reduction in the severity of both oral disease and arthritis. Moreover, FimA Ab attenuated bacterial attachment and aggregation on human gingival and rheumatoid arthritis synovial fibroblasts. In addition, we discovered bacteria may utilize dendritic cells, macrophages and neutrophils to migrate into the joints of CIA mice. These results suggest that disrupting Pg fimbriae function by FimA Ab ameliorates RA.
Collapse
Affiliation(s)
- Sang Hoon Jeong
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yoojun Nam
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Hyerin Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Juryun Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeri Alice Rim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Narae Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Kijun Lee
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Seungjin Choi
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yeonsue Jang
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Yena Kim
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji-Hoi Moon
- Department of Maxillofacial Biomedical Engineering, School of Dentistry, and Department of Life and Nanopharmaceutical Sciences, Kyung Hee University, Seoul, South Korea
| | - Seung Min Jung
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Sung-Hwan Park
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| | - Ji Hyeon Ju
- Division of Rheumatology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
15
|
Abstract
Management and therapy of rheumatoid arthritis (RA) has been revolutionized by the development and approval of the first biological disease-modifying antirheumatic drugs (bDMARDs) targeting tumor necrosis factor (TNF) α at the end of the last century. Today, numerous efficacious agents with different modes of action are available and achievement of clinical remission or, at least, low disease activity is the target of therapy. Early therapeutic interventions aiming at a defined goal of therapy (treat to target) are supposed to halt inflammation, improving symptoms and signs, and preserving structural integrity of the joints in RA. Up to now, bDMARDs approved for therapy in RA include agents with five different modes of action: TNF inhibition, T cell co-stimulation blockade, IL-6 receptor inhibition, B cell depletion, and interleukin 1 inhibition. Furthermore, targeted synthetic DMARDs (tsDMARDs) inhibiting Janus kinase (JAK) and biosimilars also are approved for RA. The present review focuses on bDMARDs and tsDMARDS regarding similarities and possible drug-specific advantages in the treatment of RA. Furthermore, compounds not yet approved in RA and biosimilars are discussed. Following the American College of Rheumatology (ACR) and European League Against Rheumatism (EULAR) recommendations, specific treatment of the disease will be discussed with respect to safety and efficacy. In particular, we discuss the question of favoring specific bDMARDs or tsDMARDs in the two settings of insufficient response to methotrexate and to the first bDMARD, respectively.
Collapse
|
16
|
Cañete JD, Hernández MV, Sanmartí R. Safety profile of biological therapies for treating rheumatoid arthritis. Expert Opin Biol Ther 2017; 17:1089-1103. [DOI: 10.1080/14712598.2017.1346078] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Affiliation(s)
- Juan D. Cañete
- Arthritis Unit, Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | - Ma Victoria Hernández
- Arthritis Unit, Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| | - Raimon Sanmartí
- Arthritis Unit, Rheumatology Department, Hospital Clinic and IDIBAPS, Barcelona, Spain
| |
Collapse
|
17
|
Na M, Wang W, Fei Y, Josefsson E, Ali A, Jin T. Both anti-TNF and CTLA4 Ig treatments attenuate the disease severity of staphylococcal dermatitis in mice. PLoS One 2017; 12:e0173492. [PMID: 28264025 PMCID: PMC5338833 DOI: 10.1371/journal.pone.0173492] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Accepted: 02/21/2017] [Indexed: 12/20/2022] Open
Abstract
Background RA patients being treated with biologics are known to have an increased risk of infections. We recently demonstrated that both CTLA4 Ig and anti-TNF treatment aggravate systemic Staphylococcus aureus (S. aureus) infection in mice, but with distinct clinical manifestations. However, the effects of CTLA4 Ig and anti-TNF treatments on a local S. aureus infection (e.g., skin infection) might differ from their effects on a systemic infection. Aims The aim of this study was to examine the differential effects of anti-TNF versus CTLA4 Ig treatment on S. aureus skin infections in mice. Method Abatacept (CTLA4 Ig), etanercept (anti-TNF treatment) or PBS was given to NMRI mice subcutaneously inoculated with S. aureus strain SH1000. The clinical signs of dermatitis, along with histopathological changes due to skin infection, were compared between the groups. Results Both CTLA4 Ig and anti-TNF treatment resulted in less severe skin infections and smaller post-infectious hyperpigmentation compared with controls. Consistent with the clinical signs of dermatitis, smaller lesion size, more epithelial hyperplasia and more granulation were found in skin biopsies from mice receiving anti-TNF compared with PBS controls. However, both CTLA4 Ig and anti-TNF therapy tended to prolong the healing time, although this finding was not statistically significant. Serum MCP-1 levels were elevated in the anti-TNF group relative to the CTLA4 Ig and PBS groups, whereas IL-6 levels were higher in PBS controls than in the other two groups. Both anti-TNF and CTLA4 Ig treatments tended to down-regulate the necrosis/apoptosis ratio in the locally infected skin tissue. Importantly, no tangible difference was found in the bacterial burden among groups. Conclusion Both CTLA4 Ig and anti-TNF therapies attenuate disease severity but may prolong the healing time required for S. aureus skin infections. Neither treatment has an impact on bacterial clearance in skin tissues.
Collapse
Affiliation(s)
- Manli Na
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at Gothenburg University, S-41346 Göteborg, Sweden
| | - Wanzhong Wang
- Department of Medical Biosciences, Umeå University, Umeå, Sweden
| | - Ying Fei
- Department of Microbiology and Immunology, The Affiliated Hospital of GuiZhou Medical University, Guiyang, P.R. China
| | - Elisabet Josefsson
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at Gothenburg University, S-41346 Göteborg, Sweden
| | - Abukar Ali
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at Gothenburg University, S-41346 Göteborg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy at Gothenburg University, S-41346 Göteborg, Sweden
- Department of Rheumatology, Sahlgrenska University Hospital, Göteborg, Sweden
- * E-mail:
| |
Collapse
|
18
|
Kawamoto H, Takasaki J, Ishii S, Suzuki M, Morino E, Naka G, Iikura M, Izumi S, Takeda Y, Sugiyama H. Re-administration of abatacept for the control of articular symptoms of rheumatoid arthritis during anti-tuberculous therapy. Respir Med Case Rep 2017; 21:147-150. [PMID: 28507894 PMCID: PMC5423351 DOI: 10.1016/j.rmcr.2017.04.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/09/2017] [Accepted: 04/10/2017] [Indexed: 11/01/2022] Open
Abstract
This case report describes the re-administration of abatacept to successfully reduce the articularsymptoms of a patient with rheumatoid arthritisduring the intensive phase of anti-tuberculous therapy. A 75-year-old man developed active pulmonary tuberculosis during the administration of abatacept for rheumatoid arthritis. The patient experienced a paradoxical reaction and exacerbation of rheumatoid arthritis that caused us to discontinue the abatacept. Later re-administration of abatacept along with anti-tuberculosis treatment led to well-controlled rheumatoid arthritis without exacerbation of the tuberculosis. This case shows that re-administration of abatacept may be much safer than TNF inhibitor to treat patients who are infected with mycobacteria during thetreatment of immunological diseases such asrheumatoid arthritiswith biological agents.
Collapse
|
19
|
Segueni N, Tritto E, Bourigault ML, Rose S, Erard F, Le Bert M, Jacobs M, Di Padova F, Stiehl DP, Moulin P, Brees D, Chibout SD, Ryffel B, Kammüller M, Quesniaux VF. Controlled Mycobacterium tuberculosis infection in mice under treatment with anti-IL-17A or IL-17F antibodies, in contrast to TNFα neutralization. Sci Rep 2016; 6:36923. [PMID: 27853279 PMCID: PMC5113257 DOI: 10.1038/srep36923] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2016] [Accepted: 10/21/2016] [Indexed: 12/20/2022] Open
Abstract
Antibodies targeting IL-17A or its receptor IL-17RA show unprecedented efficacy in the treatment of autoimmune diseases such as psoriasis. These therapies, by neutralizing critical mediators of immunity, may increase susceptibility to infections. Here, we compared the effect of antibodies neutralizing IL-17A, IL-17F or TNFα on murine host responses to Mycobacterium tuberculosis infection by evaluating lung transcriptomic, microbiological and histological analyses. Coinciding with a significant increase of mycobacterial burden and pathological changes following TNFα blockade, gene array analyses of infected lungs revealed major changes of inflammatory and immune gene expression signatures 4 weeks post-infection. Specifically, gene expression associated with host-pathogen interactions, macrophage recruitment, activation and polarization, host-antimycobacterial activities, immunomodulatory responses, as well as extracellular matrix metallopeptidases, were markedly modulated by TNFα blockade. IL-17A or IL-17F neutralization elicited only mild changes of few genes without impaired host resistance four weeks after M. tuberculosis infection. Further, the absence of both IL-17RA and IL-22 pathways in genetically deficient mice did not profoundly compromise host control of M. tuberculosis over a 6-months period, ruling out potential compensation between these two pathways, while TNFα-deficient mice succumbed rapidly. These data provide experimental confirmation of the low clinical risk of mycobacterial infection under anti-IL-17A therapy, in contrast to anti-TNFα treatment.
Collapse
Affiliation(s)
- Noria Segueni
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Elaine Tritto
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Marie-Laure Bourigault
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Stéphanie Rose
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - François Erard
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Marc Le Bert
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| | - Muazzam Jacobs
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, South Africa
- National Health Laboratory Service, Cape Town, South Africa
| | - Franco Di Padova
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Daniel P. Stiehl
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Pierre Moulin
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Dominique Brees
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Salah-Dine Chibout
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Bernhard Ryffel
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, Health Sciences Faculty, University of Cape Town, South Africa
| | - Michael Kammüller
- Novartis Institutes for Biomedical Research, CH-4002, Basel, Switzerland
| | - Valerie F. Quesniaux
- CNRS, UMR7355, Orleans, France
- University of Orleans, INEM, Experimental and Molecular Immunology and Neurogenetics, Orleans, France
| |
Collapse
|
20
|
Salmon JH, Gottenberg JE, Ravaud P, Cantagrel A, Combe B, Flipo RM, Schaeverbeke T, Houvenagel E, Gaudin P, Loeuille D, Rist S, Dougados M, Sibilia J, Le Loët X, Meyer O, Solau-Gervais E, Marcelli C, Bardin T, Pane I, Baron G, Perrodeau E, Mariette X. Predictive risk factors of serious infections in patients with rheumatoid arthritis treated with abatacept in common practice: results from the Orencia and Rheumatoid Arthritis (ORA) registry. Ann Rheum Dis 2015; 75:1108-13. [PMID: 26048170 DOI: 10.1136/annrheumdis-2015-207362] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2015] [Accepted: 05/14/2015] [Indexed: 11/04/2022]
Abstract
OBJECTIVES Little data are available regarding the rate and predicting factors of serious infections in patients with rheumatoid arthritis (RA) treated with abatacept (ABA) in daily practice. We therefore addressed this issue using real-life data from the Orencia and Rheumatoid Arthritis (ORA) registry. METHODS ORA is an independent 5-year prospective registry promoted by the French Society of Rheumatology that includes patients with RA treated with ABA. At baseline, 3 months, 6 months and every 6 months or at disease relapse, during 5 years, standardised information is prospectively collected by trained clinical nurses. A serious infection was defined as an infection occurring during treatment with ABA or during the 3 months following withdrawal of ABA without any initiation of a new biologic and requiring hospitalisation and/or intravenous antibiotics and/or resulting in death. RESULTS Baseline characteristics and comorbidities: among the 976 patients included with a follow-up of at least 3 months (total follow-up of 1903 patient-years), 78 serious infections occurred in 69 patients (4.1/100 patient-years). Predicting factors of serious infections: on univariate analysis, an older age, history of previous serious or recurrent infections, diabetes and a lower number of previous anti-tumour necrosis factor were associated with a higher risk of serious infections. On multivariate analysis, only age (HR per 10-year increase 1.44, 95% CI 1.17 to 1.76, p=0.001) and history of previous serious or recurrent infections (HR 1.94, 95% CI 1.18 to 3.20, p=0.009) were significantly associated with a higher risk of serious infections. CONCLUSIONS In common practice, patients treated with ABA had more comorbidities than in clinical trials and serious infections were slightly more frequently observed. In the ORA registry, predictive risk factors of serious infections include age and history of serious infections.
Collapse
Affiliation(s)
- J H Salmon
- Rheumatology Department, CHU Reims, Reims, France
| | - J E Gottenberg
- Rheumatology Department, National Center for Rare Systemic Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, CNRS, Institut de Biologie Moléculaire et Cellulaire, Immunopathologie et Chimie Thérapeutique/Laboratory of Excellence Medalis, Université de Strasbourg, Strasbourg, France
| | - P Ravaud
- Centre de Recherche en Epidémiologie et Statistiques, INSERM U1153, Centre d'Épidémiologie Clinique, Hôpital Hôtel-Dieu, Assistance Publique-Hôpitaux de Paris (AP-HP), Descartes University, Paris, France
| | - A Cantagrel
- Rheumatology Center, Purpan Hospital, Paul Sabatier University, Toulouse, France
| | - B Combe
- Rheumatology Department, Lapeyronie University Hospital, Montpellier I University, Montpellier, France
| | - R M Flipo
- Rheumatology Department, CHRU de Lille, Université de Lille-2, Lille, France
| | | | - E Houvenagel
- Rheumatology Department, CHU Lomme, Lomme, France
| | - P Gaudin
- Rheumatology Department, CHU Grenoble, Grenoble, France
| | - D Loeuille
- Rheumatology Department, CHU Nancy, Nancy, France
| | - S Rist
- Rheumatology Department, CHR Orléans, Orléans, France
| | - M Dougados
- Medicine Faculty, Paris-Descartes University, Paris, UPRES-EA 4058, Cochin Hospital, Rheumatology B, Paris, France
| | - J Sibilia
- Rheumatology department, National Center for Rare Systemic Autoimmune Diseases, Hôpitaux Universitaires de Strasbourg, INSERM UMRS_1109, Université de Strasbourg, Strasbourg, France
| | - X Le Loët
- Rheumatology Department, Rouen University Hospital & Inserm U905, Rouen, France
| | - O Meyer
- Rheumatology Department, Groupe Hospitalier Bichat-Claude Bernard (AP-HP), Paris, France
| | | | - C Marcelli
- Rheumatology Department, CHU Caen, Caen, France
| | - T Bardin
- Rheumatology Department, Hôpital Lariboisière, Paris, France
| | - I Pane
- Centre de Recherche en Epidémiologie et Statistiques, INSERM U1153, Centre d'Épidémiologie Clinique, Hôpital Hôtel-Dieu, Assistance Publique-Hôpitaux de Paris (AP-HP), Descartes University, Paris, France
| | - G Baron
- Centre de Recherche en Epidémiologie et Statistiques, INSERM U1153, Centre d'Épidémiologie Clinique, Hôpital Hôtel-Dieu, Assistance Publique-Hôpitaux de Paris (AP-HP), Descartes University, Paris, France
| | - E Perrodeau
- Centre de Recherche en Epidémiologie et Statistiques, INSERM U1153, Centre d'Épidémiologie Clinique, Hôpital Hôtel-Dieu, Assistance Publique-Hôpitaux de Paris (AP-HP), Descartes University, Paris, France
| | - X Mariette
- Rheumatology Department, Hôpitaux Universitaires Paris-Sud, AP-HP, INSERM U1184, IMVA: Center of Immunology of Viral Infections and Autoimmune Diseases, Paris, France
| | | |
Collapse
|
21
|
Mota LMHD, Cruz BA, Brenol CV, Pollak DF, Pinheiro GDRC, Laurindo IMM, Pereira IA, Carvalho JFD, Bertolo MB, Pinheiro MDM, Freitas MVC, Silva NAD, Louzada‐Júnior P, Sampaio‐Barros PD, Giorgi RDN, Lima RAC, Andrade LEC. Segurança do uso de terapias biológicas para o tratamento de artrite reumatoide e espondiloartrites. REVISTA BRASILEIRA DE REUMATOLOGIA 2015; 55:281-309. [DOI: 10.1016/j.rbr.2014.06.006] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 05/25/2014] [Accepted: 06/30/2014] [Indexed: 02/07/2023] Open
|
22
|
Ali A, Welin A, Schwarze JC, Svensson MND, Na M, Jarneborn A, Magnusson M, Mohammad M, Kwiecinski J, Josefsson E, Bylund J, Pullerits R, Jin T. CTLA4 Immunoglobulin but Not Anti-Tumor Necrosis Factor Therapy Promotes Staphylococcal Septic Arthritis in Mice. J Infect Dis 2015; 212:1308-16. [PMID: 25838268 DOI: 10.1093/infdis/jiv212] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2015] [Accepted: 03/25/2015] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND The development of biologics has greatly increased the quality of life and the life expectancy of many patients with rheumatoid arthritis. However, a large number of these patients have an increased risk of developing serious infections. The aim of this study was to examine differential effects of anti-tumor necrosis factor (TNF) treatment and CTLA4 immunoglobulin (Ig) treatment on both immunological response and host defense in a murine model of septic arthritis. METHODS Abatacept (CTLA4-Ig), etanercept (anti-TNF), or phosphate-buffered saline were given to NMRI mice intravenously inoculated with Staphylococcus aureus. The clinical course of septic arthritis and histopathological and radiological changes of joints were compared among the groups. RESULTS Mice receiving CTLA4-Ig treatment had more-severe septic arthritis, compared with controls and mice receiving anti-TNF treatment. Anti-TNF treatment led to more-severe weight loss and kidney abscesses, as well as a higher bacterial burden in the kidneys. Mice receiving CTLA4-Ig therapy had lower serum levels of interleukin 4, whereas mice receiving anti-TNF therapy had higher levels of TNF-α. Both iNOS and arginase-1 expression were reduced in peritoneal macrophages from mice receiving CTLA4-Ig, compared with expression in the anti-TNF group. CONCLUSIONS CTLA4-Ig therapy significantly increased the susceptibility to S. aureus septic arthritis in mice, whereas anti-TNF therapy deteriorated host bacterial clearance, resulting in more-severe weight loss and kidney abscesses.
Collapse
Affiliation(s)
- Abukar Ali
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Amanda Welin
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jan-Christoph Schwarze
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Mattias N D Svensson
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Manli Na
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Anders Jarneborn
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Malin Magnusson
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Majd Mohammad
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Jakub Kwiecinski
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Elisabet Josefsson
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Johan Bylund
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Rille Pullerits
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| | - Tao Jin
- Department of Rheumatology and Inflammation Research, Institution of Medicine, Sahlgrenska Academy, University of Gothenburg, Sweden
| |
Collapse
|
23
|
Infections and biologic therapy for rheumatoid arthritis. Rheumatology (Oxford) 2015. [DOI: 10.1016/b978-0-323-09138-1.00067-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
|
24
|
Ali A, Zhu X, Kwiecinski J, Gjertsson I, Lindholm C, Iwakura Y, Wang X, Lycke N, Josefsson E, Pullerits R, Jin T. Antibiotic-killed Staphylococcus aureus induces destructive arthritis in mice. Arthritis Rheumatol 2015; 67:107-116. [PMID: 25302691 DOI: 10.1002/art.38902] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Accepted: 09/30/2014] [Indexed: 04/15/2024]
Abstract
OBJECTIVE Permanent reduction in joint function is a severe postinfectious complication in patients with Staphylococcus aureus septic arthritis. We undertook this study to determine whether this reduction in joint function might be caused by persistent joint inflammation after the adequate eradication of bacteria by antibiotics. METHODS After intraarticular injection of cloxacillin-killed S aureus into mouse knee joints, we investigated whether antibiotic-killed S aureus induced joint inflammation and elucidated the molecular and cellular mechanisms of this type of arthritis. RESULTS Intraarticular injection of antibiotic-killed S aureus induced mild-to-moderate synovitis and bone erosions that lasted for a minimum of 14 days. Compared with wild-type animals, mice deficient in tumor necrosis factor receptor type I (TNFRI), receptor for advanced glycation end products (RAGE), or Toll-like receptor 2 (TLR-2) had a significantly reduced frequency and severity of synovitis. Combined depletion of monocytes and neutrophils also resulted in a significantly lower frequency of synovitis. Among bacterial factors, insoluble cell debris played a more important role than bacterial DNA or soluble components in inducing joint inflammation. Importantly, anti-TNF therapy abrogated joint inflammation induced by antibiotic-killed S aureus. CONCLUSION Antibiotic-killed S aureus induced and maintained joint inflammation mediated through TLR-2, TNFRI, and RAGE. The cross-talk between neutrophils and monocytes is responsible for this type of arthritis. Anti-TNF therapy might be used as a novel strategy, in combination with antibiotics, to treat staphylococcal septic arthritis.
Collapse
MESH Headings
- Animals
- Anti-Bacterial Agents/pharmacology
- Arthritis, Experimental/metabolism
- Arthritis, Experimental/microbiology
- Arthritis, Experimental/pathology
- Arthritis, Infectious/metabolism
- Arthritis, Infectious/microbiology
- Arthritis, Infectious/pathology
- Cell Communication/physiology
- Cloxacillin/pharmacology
- Disease Models, Animal
- Female
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Monocytes/pathology
- Neutrophils/pathology
- Receptor for Advanced Glycation End Products
- Receptors, Immunologic/deficiency
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- Receptors, Tumor Necrosis Factor, Type I/deficiency
- Receptors, Tumor Necrosis Factor, Type I/genetics
- Receptors, Tumor Necrosis Factor, Type I/metabolism
- Severity of Illness Index
- Staphylococcus aureus/drug effects
- Toll-Like Receptor 2/deficiency
- Toll-Like Receptor 2/genetics
- Toll-Like Receptor 2/metabolism
Collapse
Affiliation(s)
- Abukar Ali
- Sahlgrenska Academy at University of Gothenburg, Gothenburg, Sweden
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Abstract
In modern times a relationship between tuberculosis (TB) and rheumatoid arthritis (RA) has been firmly recognized, and is primarily attributable to the immunosuppressive therapies used to treat RA. Whereas TB can complicate the successful management of RA, nontuberculous mycobacteria have now perhaps become as important as (if not more so than) TB in the setting of RA, and can represent an even greater challenge to the rheumatologist wishing to use immunosuppressive therapies. This article reviews our most recent understanding of the epidemiological and clinical aspects of mycobacterial disease as it relates to RA, and the existing and emerging immunosuppressive therapies used to treat this disease.
Collapse
|
26
|
Ruderman EM. Overview of safety of non-biologic and biologic DMARDs. Rheumatology (Oxford) 2013; 51 Suppl 6:vi37-43. [PMID: 23221586 DOI: 10.1093/rheumatology/kes283] [Citation(s) in RCA: 94] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Safety data come from a number of sources. Randomized clinical trials tend to be relatively short, exclude patients with significant comorbidity, have limited numbers of subjects and are primarily powered for efficacy. The most useful post-marketing data come from large national registries, such as Britain's BSRBR, Sweden's ARTIS, Germany's RABBIT, France's DANBIO, Spain's BIODASER and North America's CORRONA. Among the most commonly used non-biologic DMARDs, MTX is associated with risks of hepatotoxicity and cytopenia, as well as pneumonitis, particularly during the first year of treatment. Regarding TNF inhibitors, there is an increased risk of infection (including serious infections) by bacterial pathogens, atypical fungi and opportunistic pathogens. When possible, pneumococcal and influenza vaccines should be given before initiation of treatment with any biologic DMARD. Screening for latent tuberculosis is recommended for all TNF inhibitors, and has been shown to reduce the risk of reactivation. Evidence from registries suggests that there is no increased risk of solid tumours with TNF inhibitor treatment; however, non-melanoma skin cancers are more common. Specific risks with other biologic DMARDs include gastrointestinal perforation with tocilizumab, progressive multifocal leucoencephalopathy with rituximab and pulmonary infections with abatacept. Overall, the safety of biologic and non-biologic DMARDs appears to be reasonable, particularly compared with the risks associated with the disease itself.
Collapse
Affiliation(s)
- Eric M Ruderman
- Department of Medicine - Rheumatology Division, Northwestern University Feinberg School of Medicine, 675 North St Clair, Suite 14-100, Chicago, IL 60611, USA.
| |
Collapse
|
27
|
Abstract
Tuberculosis (TB) develops in 5% to 10% of people infected with Mycobacterium tuberculosis (M.tb), but we do not understand how TB develops. CBA/J mice may model these events, as sick mice share features with TB patients, including weight loss, M.tb growth, extensive granulomatous infiltrates, neutrophils, necrosis, and fibrosis. Here, M.tb-infected CBA/J mice were categorized clinically: those with no signs or those with 10% weight loss to determine whether clinical state was associated with lung lesions. The type and distribution of infiltrates (granulomatous with lymphoid aggregates and scattered neutrophils) were similar in mice with weight loss and in mice with no signs. The amount of infiltration and neutrophil foci were higher in mice with weight loss than in mice with no clinical signs. Necrosis and fibrosis were only identified in mice that lost weight. Our results suggest that CBA/J mice may be useful to determine if and how neutrophils contribute to TB disease progression in mouse models.
Collapse
Affiliation(s)
- S Major
- Department of Infectious Disease and Global Health, Tufts University, 200 Westboro Rd, Bldg 20, Grafton, MA 01536, USA.
| | | | | |
Collapse
|
28
|
Winthrop KL. Infections and biologic therapy in rheumatoid arthritis: our changing understanding of risk and prevention. Rheum Dis Clin North Am 2012; 38:727-45. [PMID: 23137579 DOI: 10.1016/j.rdc.2012.08.019] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Patients with rheumatoid arthritis are at higher risk for serious infections and death from infection than the general public. Prednisone and biologic agents increase this risk, although the risk associated with biologics can be mitigated when such agents act as prednisone-sparing therapies. Some of the important causes of infectious morbidity in this setting are preventable with screening (eg, tuberculosis) or vaccination (eg, herpes zoster).
Collapse
Affiliation(s)
- Kevin L Winthrop
- Public Health and Preventive Medicine, Oregon Health & Science University, Portland, OR, USA.
| |
Collapse
|
29
|
Martín Mola E, Balsa A, Martínez Taboada V, Sanmartí R, Marenco JL, Navarro Sarabia F, Gómez-Reino J, Alvaro-Gracia JM, Román Ivorra JA, Lojo L, Plasencia C, Carmona L. Abatacept use in rheumatoid arthritis: evidence review and recommendations. ACTA ACUST UNITED AC 2012; 9:5-17. [PMID: 22766432 DOI: 10.1016/j.reuma.2012.05.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2012] [Revised: 04/29/2012] [Accepted: 05/02/2012] [Indexed: 11/16/2022]
Abstract
OBJECTIVE To review the clinical evidence on abatacept and to formulate recommendations in order to clear up points related to its use in rheumatology. METHOD An expert panel of rheumatologists objectively summarized the evidence on the mechanism of action, practicalities, effectiveness and safety of abatacept, and formulated recommendations following a literature review. The level of evidence and degree of recommendation was established. RESULTS The document presents 21 statements focused on evidence or recommendations on abatacept (14 evidence summaries and 9 recommendations). The level of evidence was 2b or higher according to the Oxford Centre for Evidence-Based Medicine scale on 14 occasions. The degree of the recommendation was A in two recommendations, C in one, and D in the rest. It was considered important to make recommendations on aspects with lower levels of evidence. CONCLUSIONS This is a practical document to supplement the summary of product characteristics.
Collapse
Affiliation(s)
- Emilio Martín Mola
- Servicio de Reumatología, Hospital Universitario La Paz, IdiPAZ, Madrid, España.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Bugelski PJ, Martin PL. Concordance of preclinical and clinical pharmacology and toxicology of therapeutic monoclonal antibodies and fusion proteins: cell surface targets. Br J Pharmacol 2012; 166:823-46. [PMID: 22168282 PMCID: PMC3417412 DOI: 10.1111/j.1476-5381.2011.01811.x] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2011] [Revised: 10/14/2011] [Accepted: 11/28/2011] [Indexed: 12/20/2022] Open
Abstract
Monoclonal antibodies (mAbs) and fusion proteins directed towards cell surface targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 15 currently approved mAbs and fusion proteins targeted to the cell surface. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions'; and the US Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the 15 approved biopharmaceuticals were included: abatacept; abciximab; alefacept; alemtuzumab; basiliximab; cetuximab; daclizumab; efalizumab; ipilimumab; muromonab; natalizumab; panitumumab; rituximab; tocilizumab; and trastuzumab. For statistical analysis of concordance, data from these 15 were combined with data on the approved mAbs and fusion proteins directed towards soluble targets. Good concordance with human pharmacodynamics was found for mice receiving surrogates or non-human primates (NHPs) receiving the human pharmaceutical. In contrast, there was poor concordance for human pharmacodynamics in genetically deficient mice and for human adverse effects in all three test systems. No evidence that NHPs have superior predictive value was found.
Collapse
Affiliation(s)
- Peter J Bugelski
- Biologics Toxicology, Janssen Research & Development, division of Johnson & Johnson Pharmaceutical Research & Development, LLC, Radnor, PA 19087, USA
| | | |
Collapse
|
31
|
Pham T, Bachelez H, Berthelot JM, Blacher J, Claudepierre P, Constantin A, Fautrel B, Gaujoux-Viala C, Goëb V, Gossec L, Goupille P, Guillaume-Czitrom S, Hachulla E, Lequerré T, Marolleau JP, Martinez V, Masson C, Mouthon L, Puéchal X, Richette P, Saraux A, Schaeverbeke T, Soubrier M, Viguier M, Vittecoq O, Wendling D, Mariette X, Sibilia J. Abatacept therapy and safety management. Joint Bone Spine 2012; 79 Suppl 1:3-84. [DOI: 10.1016/s1297-319x(12)70011-8] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
32
|
Herrero-Beaumont G, Martínez Calatrava MJ, Castañeda S. Mecanismo de acción de abatacept: concordancia con su perfil clínico. ACTA ACUST UNITED AC 2012; 8:78-83. [DOI: 10.1016/j.reuma.2011.08.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2011] [Revised: 07/30/2011] [Accepted: 08/24/2011] [Indexed: 01/27/2023]
|
33
|
Franco NH, Correia-Neves M, Olsson IAS. How "humane" is your endpoint? Refining the science-driven approach for termination of animal studies of chronic infection. PLoS Pathog 2012; 8:e1002399. [PMID: 22275862 PMCID: PMC3261900 DOI: 10.1371/journal.ppat.1002399] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Affiliation(s)
- Nuno H. Franco
- IBMC - Institute for Molecular and Cell Biology (Laboratory Animal Science Group), University of Porto, Porto, Portugal
- * E-mail:
| | - Margarida Correia-Neves
- Life and Health Sciences Research Institute (ICVS), School of Health Sciences, University of Minho, Braga, Portugal
- ICVS/3B’s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - I. Anna S. Olsson
- IBMC - Institute for Molecular and Cell Biology (Laboratory Animal Science Group), University of Porto, Porto, Portugal
| |
Collapse
|
34
|
|
35
|
Keyser FD. Choice of Biologic Therapy for Patients with Rheumatoid Arthritis: The Infection Perspective. Curr Rheumatol Rev 2011; 7:77-87. [PMID: 22081766 PMCID: PMC3182090 DOI: 10.2174/157339711794474620] [Citation(s) in RCA: 70] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 07/30/2010] [Accepted: 11/10/2010] [Indexed: 12/27/2022]
Abstract
Biologicals revolutionized the treatment of Rheumatoid Arthritis (RA). The targeted suppression of key inflammatory pathways involved in joint inflammation and destruction allows better disease control, which, however, comes at the price of an elevated infection risk due to relative immunosuppression. The disease-related infection risk and the infection risk associated with the use of TNF-α inhibitors (infliximab, adalimumab, etanercept, golimumab and certolizumab pegol), rituximab, abatacept and tocilizumab are discussed. Risk factors clinicians need to take into account when selecting the most appropriate biologic therapy for RA patients, as well as precautions and screening concerning a number of specific infections, such as tuberculosis, intracellular bacterial infections, reactivation of chronic viral infections and HIV are reviewed.
Collapse
|
36
|
Infectious Complications Associated with Immunomodulating Biologic Agents. Hematol Oncol Clin North Am 2011; 25:117-38. [DOI: 10.1016/j.hoc.2010.11.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
37
|
Dhawale R, Moreland LW. T-cell co-stimulation. Rheumatology (Oxford) 2011. [DOI: 10.1016/b978-0-323-06551-1.00058-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|
38
|
Winthrop KL. Infections and biologic therapy in rheumatoid arthritis. Rheumatology (Oxford) 2011. [DOI: 10.1016/b978-0-323-06551-1.00064-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
39
|
Inhibition of T cells provides protection against early invasive pneumococcal disease. Infect Immun 2010; 78:5287-94. [PMID: 20855509 DOI: 10.1128/iai.00431-10] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Infections caused by Streptococcus pneumoniae are major causes of morbidity and mortality, which are in part mediated by immune cell-dependent mechanisms. Yet, the specific contributions of individual cell types to immunopathology are only partially understood. T cells are well characterized with respect to their function in protective humoral immune responses; however, their roles during early stages of infection and invasive pneumococcal disease (IPD) are less well defined. Using a mouse model of pneumococcal sepsis, we found that CD4(+) T cells were recruited to the lung as early as 12 h after intranasal infection. Recruitment was accompanied by upregulation of CD69 and B7-H1, reflecting T-cell activation. Unexpectedly, major histocompatibility complex (MHC) class II-deficient mice, which lack CD4(+) T cells, displayed an increased survival despite comparable bacterial titers in the blood, spleen, and lung. The higher survival correlated with a lower cytokine and chemokine response upon S. pneumoniae challenge in MHC class II-deficient mice, suggesting that inflammation may contribute to the mortality of IPD. Comparable to the case for MHC class II-deficient mice, antibody-mediated depletion of CD4(+) T cells and drug-induced inhibition of T-cell function with cyclosporine, or interference with T-cell activation using CTLA4-immunoglobulin (Abatacept), led to significant increases in survival during IPD. Our results reveal an important and adverse role of CD4(+) T cells in the pathogenesis of IPD and suggest that modulation of T-cell activation during early phases of S. pneumoniae invasive infection may provide a therapeutic option.
Collapse
|
40
|
|
41
|
Posadas A, Lisse J, Sarkar S. Abatacept in the treatment of rheumatoid arthritis. Expert Rev Clin Immunol 2010; 5:9-17. [PMID: 20476895 DOI: 10.1586/1744666x.5.1.9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Rheumatoid arthritis (RA) is a chronic inflammatory arthritis affecting 1% of the population. The immunologic dysfunction underlying this immune disorder is complex and intricate with the involvement of various immune cells as well as cytokines and surface molecules. While inhibition of TNF-alpha has changed the outlook of patients with this disorder, it regulates only one aspect of the inflammatory cascade associated with RA. This is corroborated by experience in the clinic, where a significant proportion of the patients do not have clinical benefit with such therapies. Furthermore, a number of patients experience blunting of the initial therapeutic benefits of TNF-alpha-targeted therapies. Thus, a different approach to regulate the immune dysfunction associated with RA is necessary. T cells are considered important in the pathogenesis of RA and abatacept, a fusion protein, was developed to abolish the activation of the T cell by blocking its interaction with the antigen-presenting cell. Abatacept has demonstrated promising clinical improvements in patients with RA. Although clinical experience with this new drug is limited and its mechanism of action remains to be understood, the data on the safety profile are reassuring.
Collapse
Affiliation(s)
- Augusto Posadas
- Section of Rheumatology, Department of Medicine, University of Arizona, Tucson, AZ, USA
| | | | | |
Collapse
|
42
|
Koo S, Marty FM, Baden LR. Infectious Complications Associated with Immunomodulating Biologic Agents. Infect Dis Clin North Am 2010; 24:285-306. [DOI: 10.1016/j.idc.2010.01.006] [Citation(s) in RCA: 65] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
43
|
Abstract
PURPOSE OF REVIEW Although the remarkable efficacy of biological therapy has resulted in significant success in rheumatic disease management, susceptibility to infections remains a concern. Here we review the latest publications on infectious complications of biological therapy in rheumatic diseases. RECENT FINDINGS The recent data on anti-tumor necrosis factor agents show encouraging results in relation to infections. The majority of the infections are minor, and opportunistic infections including tuberculosis are rare. The incidence of infections decreases with time on biologic therapy. Vaccination is effective while on biological agents, although live vaccines should be avoided. Biologic therapy in the setting of HIV, HCV and HBV continues to be studied, but data are accumulating in support of a favorable safety profile. There are degrees of differential susceptibility to infection across the rheumatic diseases, which should be taken into account in weighing the infectious risks of biologics in the respective diseases. SUMMARY Biological medications have a favorable safety profile but continued vigilance is appropriate. Most infectious reported episodes are minor and the risk of infection appears to decrease with duration of treatment.
Collapse
|
44
|
Acevedo-Vásquez E, Ponce de León D, Gamboa-Cárdenas R. Latent infection and tuberculosis disease in rheumatoid arthritis patients. Rheum Dis Clin North Am 2009; 35:163-81. [PMID: 19481003 DOI: 10.1016/j.rdc.2009.03.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
New drug classes, biologics, have been developed over the past 10 years based on human or chimeric antibodies against cytokines or receptors with pivotal roles in the inflammatory pathways of immune-mediated inflammatory disease. Anti-tumor necrosis factor agents carry the largest infection risk of all the biologics, predisposing patients to mycobacterial infections. Patients receiving biologics are at higher risk for developing tuberculosis. New cases of tuberculosis or reactivation of latent tuberculosis infections may occur during the course of treatment, so a high level of vigilance is highly recommended.
Collapse
|
45
|
Lutt JR. Efficacy, safety, and tolerability of abatacept in the management of rheumatoid arthritis. Open Access Rheumatol 2009; 1:17-35. [PMID: 27789979 PMCID: PMC5074723 DOI: 10.2147/oarrr.s4536] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The management of rheumatoid arthritis (RA) has undergone an impressive transformation over the past few decades. Further understanding of the pathophysiology of the disease process has resulted in the development of biologic agents that target proinflammatory cytokines and both B and T lymphocytes. By blocking an important costimulatory pathway, abatacept leads to a dramatic reduction in T cell stimulation and proliferation. Multiple clinical trials have revealed consistent benefit with regards to clinical and radiographic efficacy, quality of life, and disability in patients suffering from RA who have had inadequate responses to methotrexate or tumor necrosis factor inhibitors. The possibility of remission when used early in the disease course has also been demonstrated. Importantly, abatacept has been very well tolerated with a low rate of serious infections and no apparent increase in malignancies to date. Continued surveillance of the benefits and risks will help to better define its place amongst the other biologic agents in the treatment of RA.
Collapse
Affiliation(s)
- Joseph R Lutt
- Division of Arthritis and Rheumatic Diseases, Oregon Health and Science University, Portland, Oregon, USA
| |
Collapse
|
46
|
Bhatt K, Uzelac A, Mathur S, McBride A, Potian J, Salgame P. B7 costimulation is critical for host control of chronic Mycobacterium tuberculosis infection. THE JOURNAL OF IMMUNOLOGY 2009; 182:3793-800. [PMID: 19265158 DOI: 10.4049/jimmunol.0802996] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Although much is understood regarding the role of B7/CD28 family of costimulatory molecules in regulating host resistance in the context of several pathogens, analogous information with Mycobacterium tuberculosis is lacking. To address the requirements of B7-mediated costimulation in host resistance against tuberculosis, mice deficient in both B7.1 and B7.2 (B7DKO) were aerosol infected with M. tuberculosis Erdman and disease progression was monitored. We report herein that B7DKO mice are initially able to contain the bacterial load in the lung, but exhibit enhanced susceptibility during chronic infection. Despite the early control of bacterial replication, B7DKO mice essentially start off with compromised Th1 immunity and slower granulomatous response in the lung, characterized by markedly reduced lymphocytic infiltration. As the infection progresses from acute phase to the chronic phase, the nascent granulomas in the B7DKO lungs never fully achieve the architecture of granulomas developing in wild-type mice. Instead, lesions spread progressively to involve much of the lung in the B7DKO mice, ultimately leading to necrosis. Thus, early control of M. tuberculosis growth in the lung can occur in the absence of B7 costimulation and is less dependent on Th1 immunity and formation of a granulomatous structure. However, B7 costimulation is critical for long-term containment of infection within lung granulomas. These findings suggest that the use of costimulation-based immunomodulators may have significant repercussions on the induction of host protective immunity against tuberculosis.
Collapse
Affiliation(s)
- Kamlesh Bhatt
- Department of Medicine, University of Medicine and Dentistry of New Jersey, Center for Emerging Pathogens, Newark, NJ 07101, USA
| | | | | | | | | | | |
Collapse
|
47
|
|
48
|
Dejaco C, Duftner C, Wipfler E, Schirmer M. Neue Option in der Behandlung der rheumatoiden Arthritis: Abatacept, ein selektiver Modulator der T-Zell-Kostimulation. Wien Med Wochenschr 2009; 159:70-5. [DOI: 10.1007/s10354-008-0590-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Accepted: 07/21/2008] [Indexed: 01/01/2023]
|
49
|
Furst DE. The risk of infections with biologic therapies for rheumatoid arthritis. Semin Arthritis Rheum 2008; 39:327-46. [PMID: 19117595 DOI: 10.1016/j.semarthrit.2008.10.002] [Citation(s) in RCA: 184] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2008] [Revised: 09/09/2008] [Accepted: 10/01/2008] [Indexed: 01/06/2023]
Abstract
OBJECTIVES To assess the risk of serious and nonserious bacterial and viral infections associated with the use of biologic therapy (abatacept, adalimumab, anakinra, etanercept, infliximab, and rituximab) in patients with rheumatoid arthritis (RA). METHODS Information was derived from PubMed, EMBASE, and the Cochrane clinical trials register and database of systematic reviews and relevant congress abstracts up to and including February 2008. RESULTS Compared with the general population, patients with RA have a heightened risk of infection, including tuberculosis. Long-term clinical trials and postmarketing studies indicate that anakinra and the tumor necrosis factor (TNF) inhibitors are associated with an increased risk of infections versus conventional disease-modifying antirheumatic drugs (DMARDs), especially early in the course of treatment. The most common sites of infection are the respiratory tract (including pneumonia), skin and soft tissue, and the urinary tract. The risk of tuberculosis also appears higher with TNF inhibitors (in particular, infliximab) versus DMARDs, although this can be reduced by screening and prophylaxis. TNF inhibitors do not appear to significantly increase the risk of reactivating chronic viral infections. Influenza and pneumococcal vaccinations are generally effective in the face of TNF inhibitors or abatacept. Available data suggest that the risk of infections and serious infections with abatacept and rituximab may be similar to that of the TNF inhibitors. To date, there have been no reports from clinical trials of increased tuberculosis or opportunistic infections with abatacept or rituximab. CONCLUSIONS All marketed TNF inhibitors for compared to control RA appear to increase the risk of serious and nonserious infections compared with DMARDs. Although suggestive, data for abatacept and rituximab are less definitive and longer periods of patient exposure to these agents are needed before an assessment of their risks can be made.
Collapse
Affiliation(s)
- Daniel E Furst
- University of California Los Angeles, Los Angeles, CA 90095-1670, USA.
| |
Collapse
|
50
|
Current World Literature. Curr Opin Allergy Clin Immunol 2008; 8:590-3. [DOI: 10.1097/aci.0b013e32831ceb82] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|