1
|
Samanta A, Srivastava A. Biologics in the management of pediatric inflammatory bowel disease: When and what to choose. World J Clin Pediatr 2025; 14:100938. [PMID: 40059900 PMCID: PMC11686582 DOI: 10.5409/wjcp.v14.i1.100938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/14/2024] [Accepted: 12/02/2024] [Indexed: 12/20/2024] Open
Abstract
Pediatric inflammatory bowel disease (PIBD) is a chronic inflammatory disorder of the gastrointestinal tract, with rising global incidence and prevalence. Over the past two decades, biologics have added to the therapeutic armamentarium and revolutionized the approach to treatment of inflammatory bowel disease. The available biologics include monoclonal antibodies which target inflammatory cytokines (anti-tumor necrosis factor alpha, anti-interleukin 12/23) or recruitment of leucocytes to the gastrointestinal tract (anti-alpha4beta7 integrin) and small molecules (Janus kinase inhibitors, sphingosine 1-phosphate-inhibitors) which modify the proinflammatory signaling. Considering their potential disease-modifying ability, recent pediatric guidelines from the West have advocated upfront use of biologics in appropriate clinical scenarios as a top-down approach rather than the conventional step-up approach. Although real-world studies are available regarding the clinical efficacy of biologics in PIBD, there is paucity of long-term outcome and safety data in children. Also, little information is available about the best approach in the newly industrialized - developing countries where PIBD is rising but at the same time, infections are prevalent and resources are limited. In this review, we summarize the efficacy and safety profile of biologics and small molecule drugs and discuss the challenges in the management of PIBD, especially in the developing world, and future directions.
Collapse
Affiliation(s)
- Arghya Samanta
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| | - Anshu Srivastava
- Department of Pediatric Gastroenterology, Sanjay Gandhi Postgraduate Institute of Medical Sciences, Lucknow 226014, Uttar Pradesh, India
| |
Collapse
|
2
|
Matran R, Diaconu AM, Iordache AM, Dijmărescu I, Coroleucă A, Păcurar D, Becheanu C. Anti-Tumor Necrosis Factor-α Use in Pediatric Inflammatory Bowel Disease-Reports from a Romanian Center. Pharmaceuticals (Basel) 2025; 18:84. [PMID: 39861147 PMCID: PMC11768541 DOI: 10.3390/ph18010084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Background/Objectives: The introduction of anti-tumor necrosis factor-α (anti-TNF-α) agents, particularly infliximab (IFX) and adalimumab (ADA), has significantly expanded the therapeutic arsenal for inflammatory bowel disease (IBD). While these biologics have demonstrated substantial efficacy, they are associated with a spectrum of potential adverse events (AEs). This study aims to evaluate and document these AEs to facilitate optimal patient selection and monitoring strategies of patients undergoing these therapies. Methods: This retrospective, single-center study examined pediatric IBD patients receiving anti-TNF-α therapy at the "Grigore Alexandrescu" Emergency Hospital for Children in Bucharest, Romania, from January 2015 to October 2024. AEs were categorized into non-infectious complications (acute infusion reactions, anti-drug antibody formation), dermatological effects (erythema nodosum, vasculitis), neurological effects (Guillain-Barré syndrome), and infections. AEs were analyzed in relation to the specific anti-TNF-α agent administered and comprehensively characterized. Results: Of 40 patients enrolled, 22 (55%) had Crohn's disease (CD). The median (IQR) age at diagnosis was 14.8 years [10.8-15.9]. IFX was used in 34 (85%) patients while 6 (15%) patients received either ADA or IFX/ADA sequential therapy. Twenty-seven AEs were documented in 19 (47.5%) patients, the most prevalent being antidrug antibody formation (44.4%), infections (22.2%), and acute infusion reactions (22.2%). All ADA-exposed patients experienced at least one AE, compared to 41.2% (n = 14) patients treated with IFX, p = 0.01. Conclusions: AEs were observed in approximately half of the study cohort, with anti-drug antibody formation emerging as the most frequent complication. ADA therapy was associated with a significantly higher rate of AEs compared to IFX. These findings underscore the critical importance of vigilant monitoring for patients undergoing anti-TNF-α therapy in pediatric IBD management.
Collapse
Affiliation(s)
- Roxana Matran
- Department of Paediatrics, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.M.); (I.D.); (A.C.); (D.P.); (C.B.)
- “Grigore Alexandrescu” Emergency Hospital for Children, 011743 Bucharest, Romania
| | | | | | - Irina Dijmărescu
- Department of Paediatrics, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.M.); (I.D.); (A.C.); (D.P.); (C.B.)
- “Grigore Alexandrescu” Emergency Hospital for Children, 011743 Bucharest, Romania
| | - Alexandra Coroleucă
- Department of Paediatrics, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.M.); (I.D.); (A.C.); (D.P.); (C.B.)
- “Grigore Alexandrescu” Emergency Hospital for Children, 011743 Bucharest, Romania
| | - Daniela Păcurar
- Department of Paediatrics, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.M.); (I.D.); (A.C.); (D.P.); (C.B.)
- “Grigore Alexandrescu” Emergency Hospital for Children, 011743 Bucharest, Romania
| | - Cristina Becheanu
- Department of Paediatrics, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania; (R.M.); (I.D.); (A.C.); (D.P.); (C.B.)
- “Grigore Alexandrescu” Emergency Hospital for Children, 011743 Bucharest, Romania
| |
Collapse
|
3
|
Zong Y, Kamoi K, Miyagaki M, Zhang J, Yang M, Zou Y, Ohno-Matsui K. Applications of Biological Therapy for Latent Infections: Benefits and Risks. Int J Mol Sci 2024; 25:9184. [PMID: 39273134 PMCID: PMC11394918 DOI: 10.3390/ijms25179184] [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: 07/18/2024] [Revised: 08/20/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Biological therapies have revolutionized medical treatment by targeting the key mediators or receptors involved in inflammatory responses, thereby effectively suppressing inflammation and achieving beneficial outcomes. They are more advanced than conventional therapies using corticosteroids and immunosuppressants, offering effective solutions for autoimmune diseases, cancer, transplant rejection, and various infectious diseases, including coronavirus disease 2019. Although they exert low immunosuppressive effects, biological therapies can reactivate specific biological targets associated with infections. This review summarizes the currently available biological therapies and discusses their immunosuppressive mechanisms and clinical applications, highlighting the variations in the types and frequencies of infection recurrence induced by different biological agents. Additionally, this review describes the risk factors associated with various biological agents, thus aiding clinicians in selecting the most appropriate biological therapy.
Collapse
Affiliation(s)
- Yuan Zong
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Koju Kamoi
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Miki Miyagaki
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Jing Zhang
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Mingming Yang
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Yaru Zou
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| | - Kyoko Ohno-Matsui
- Department of Ophthalmology & Visual Science, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo 113-8510, Japan
| |
Collapse
|
4
|
Kim ES, Kang B. Infliximab vs adalimumab: Points to consider when selecting anti-tumor necrosis factor agents in pediatric patients with Crohn's disease. World J Gastroenterol 2023; 29:2784-2797. [PMID: 37274072 PMCID: PMC10237103 DOI: 10.3748/wjg.v29.i18.2784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 03/20/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
Biologic agents with various mechanisms against Crohn's disease (CD) have been released and are widely used in clinical practice. However, two anti-tumor necrosis factor (TNF) agents, infliximab (IFX) and adalimumab (ADL), are the only biologic agents approved by the Food and Drug Administration for pediatric CD currently. Therefore, in pediatric CD, the choice of biologic agents should be made more carefully to achieve the therapeutic goal. There are currently no head-to-head trials of biologic agents in pediatric or adult CD. There is a lack of accumulated data for pediatric CD, which requires the extrapolation of adult data for the positioning of biologics in pediatric CD. From a pharmacokinetic point of view, IFX is more advantageous than ADL when the inflammatory burden is high, and ADL is expected to be advantageous over IFX in sustaining remission in the maintenance phase. Additionally, we reviewed the safety profile, immunogenicity, preference, and compliance between IFX and ADL and provide practical insights into the choice of anti-TNF therapy in pediatric CD. Careful evaluation of clinical indications and disease behavior is essential when prescribing anti-TNF agents. In addition, factors such as the efficacy of induction and maintenance of remission, safety profile, immunogenicity, patient preference, and compliance play an important role in evaluating and selecting treatment options.
Collapse
Affiliation(s)
- Eun Sil Kim
- Department of Pediatrics, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul 03181, South Korea
| | - Ben Kang
- Department of Pediatrics, School of Medicine, Kyungpook National University, Daegu 41944, South Korea
| |
Collapse
|
5
|
Dos Santos Nascimento IJ, da Silva-Júnior EF. TNF-α Inhibitors from Natural Compounds: An Overview, CADD Approaches, and their Exploration for Anti-inflammatory Agents. Comb Chem High Throughput Screen 2022; 25:2317-2340. [PMID: 34269666 DOI: 10.2174/1386207324666210715165943] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 05/31/2021] [Accepted: 06/01/2021] [Indexed: 02/07/2023]
Abstract
Inflammation is a natural process that occurs in the organism in response to harmful external agents. Despite being considered beneficial, exaggerated cases can cause severe problems for the body. The main inflammatory manifestations are pain, increased temperature, edema, decreased mobility, and quality of life for affected individuals. Diseases such as arthritis, cancer, allergies, infections, arteriosclerosis, neurodegenerative diseases, and metabolic problems are mainly characterized by an exaggerated inflammatory response. Inflammation is related to two categories of substances: pro- and anti-inflammatory mediators. Among the pro-inflammatory mediators is Tumor Necrosis Factor-α (TNF-α). It is associated with immune diseases, cancer, and psychiatric disorders which increase its excretion. Thus, it becomes a target widely used in discovering new antiinflammatory drugs. In this context, secondary metabolites biosynthesized by plants have been used for thousands of years and continue to be one of the primary sources of new drug scaffolds against inflammatory diseases. To decrease costs related to the drug discovery process, Computer-Aided Drug Design (CADD) techniques are broadly explored to increase the chances of success. In this review, the main natural compounds derived from alkaloids, flavonoids, terpene, and polyphenols as promising TNF-α inhibitors will be discussed. Finally, we applied a molecular modeling protocol involving all compounds described here, suggesting that their interactions with Tyr59, Tyr119, Tyr151, Leu57, and Gly121 residues are essential for the activity. Such findings can be useful for research groups worldwide to design new anti-inflammatory TNF-α inhibitors.
Collapse
Affiliation(s)
| | - Edeildo Ferreira da Silva-Júnior
- Institute of Chemistry and Biotechnology, Federal University of Alagoas, Maceió, Brazil.,Laboratory of Medicinal Chemistry, Pharmaceutical Sciences Institute, Federal University of Alagoas, Maceió, Brazil
| |
Collapse
|
6
|
Ellwanger JH, Fearnside PM, Ziliotto M, Valverde-Villegas JM, Veiga ABGDA, Vieira GF, Bach E, Cardoso JC, Müller NFD, Lopes G, Caesar L, Kulmann-Leal B, Kaminski VL, Silveira ES, Spilki FR, Weber MN, Almeida SEDEM, Hora VPDA, Chies JAB. Synthesizing the connections between environmental disturbances and zoonotic spillover. AN ACAD BRAS CIENC 2022; 94:e20211530. [PMID: 36169531 DOI: 10.1590/0001-3765202220211530] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 03/03/2022] [Indexed: 11/22/2022] Open
Abstract
Zoonotic spillover is a phenomenon characterized by the transfer of pathogens between different animal species. Most human emerging infectious diseases originate from non-human animals, and human-related environmental disturbances are the driving forces of the emergence of new human pathogens. Synthesizing the sequence of basic events involved in the emergence of new human pathogens is important for guiding the understanding, identification, and description of key aspects of human activities that can be changed to prevent new outbreaks, epidemics, and pandemics. This review synthesizes the connections between environmental disturbances and increased risk of spillover events based on the One Health perspective. Anthropogenic disturbances in the environment (e.g., deforestation, habitat fragmentation, biodiversity loss, wildlife exploitation) lead to changes in ecological niches, reduction of the dilution effect, increased contact between humans and other animals, changes in the incidence and load of pathogens in animal populations, and alterations in the abiotic factors of landscapes. These phenomena can increase the risk of spillover events and, potentially, facilitate new infectious disease outbreaks. Using Brazil as a study model, this review brings a discussion concerning anthropogenic activities in the Amazon region and their potential impacts on spillover risk and spread of emerging diseases in this region.
Collapse
Affiliation(s)
- Joel Henrique Ellwanger
- Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunobiologia e Imunogenética, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil
| | - Philip Martin Fearnside
- Instituto Nacional de Pesquisas da Amazônia/INPA, Avenida André Araújo, 2936, Aleixo, 69067-375 Manaus, AM, Brazil
| | - Marina Ziliotto
- Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunobiologia e Imunogenética, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil
| | - Jacqueline María Valverde-Villegas
- Institut de Génétique Moléculaire de Montpellier/IGMM, Centre National de la Recherche Scientifique/CNRS, Laboratoire coopératif IGMM/ABIVAX, 1919, route de Mende, 34090 Montpellier, Montpellier, France
| | - Ana Beatriz G DA Veiga
- Universidade Federal de Ciências da Saúde de Porto Alegre/UFCSPA, Departamento de Ciências Básicas de Saúde, Rua Sarmento Leite, 245, Centro Histórico, 90050-170 Porto Alegre, RS, Brazil
| | - Gustavo F Vieira
- Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunoinformática, Núcleo de Bioinformática do Laboratório de Imunogenética/NBLI, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Saúde e Desenvolvimento Humano, Universidade La Salle, Laboratório de Saúde Humana in silico, Avenida Victor Barreto, 2288, Centro, 92010-000 Canoas, RS, Brazil
| | - Evelise Bach
- Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunobiologia e Imunogenética, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil
| | - Jáder C Cardoso
- Centro Estadual de Vigilância em Saúde/CEVS, Divisão de Vigilância Ambiental em Saúde, Secretaria da Saúde do Estado do Rio Grande do Sul, Avenida Ipiranga, 5400, Jardim Botânico, 90610-000 Porto Alegre, RS, Brazil
| | - Nícolas Felipe D Müller
- Centro Estadual de Vigilância em Saúde/CEVS, Divisão de Vigilância Ambiental em Saúde, Secretaria da Saúde do Estado do Rio Grande do Sul, Avenida Ipiranga, 5400, Jardim Botânico, 90610-000 Porto Alegre, RS, Brazil
| | - Gabriel Lopes
- Fundação Oswaldo Cruz/FIOCRUZ, Casa de Oswaldo Cruz, Avenida Brasil, 4365, Manguinhos, 21040-900 Rio de Janeiro, RJ, Brazil
| | - Lílian Caesar
- Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Indiana University/IU, Department of Biology, 915 East 3rd Street, Bloomington, IN 47405, USA
| | - Bruna Kulmann-Leal
- Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunobiologia e Imunogenética, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil
| | - Valéria L Kaminski
- Programa de Pós-Graduação em Biotecnologia, Universidade Federal de São Paulo/UNIFESP, Instituto de Ciência e Tecnologia/ICT, Laboratório de Imunologia Aplicada, Rua Talim, 330, Vila Nair, 12231-280 São José dos Campos, SP, Brazil
| | - Etiele S Silveira
- Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunoinformática, Núcleo de Bioinformática do Laboratório de Imunogenética/NBLI, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil
| | - Fernando R Spilki
- Universidade Feevale, Laboratório de Saúde Única, Instituto de Ciências da Saúde/ICS, Rodovia ERS-239, 2755, Vila Nova, 93525-075 Novo Hamburgo, RS, Brazil
| | - Matheus N Weber
- Universidade Feevale, Laboratório de Saúde Única, Instituto de Ciências da Saúde/ICS, Rodovia ERS-239, 2755, Vila Nova, 93525-075 Novo Hamburgo, RS, Brazil
| | - Sabrina E DE Matos Almeida
- Universidade Feevale, Laboratório de Saúde Única, Instituto de Ciências da Saúde/ICS, Rodovia ERS-239, 2755, Vila Nova, 93525-075 Novo Hamburgo, RS, Brazil
| | - Vanusa P DA Hora
- Programa de Pós-Graduação em Ciências da Saúde, Universidade Federal do Rio Grande/FURG, Faculdade de Medicina, Rua Visconde de Paranaguá, 102, Centro, 96203-900, Rio Grande, RS, Brazil
| | - José Artur B Chies
- Universidade Federal do Rio Grande do Sul/UFRGS, Laboratório de Imunobiologia e Imunogenética, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil.,Programa de Pós-Graduação em Genética e Biologia Molecular/PPGBM, Universidade Federal do Rio Grande do Sul/UFRGS, Departmento de Genética, Campus do Vale, Avenida Bento Gonçalves, 9500, Agronomia, 91501-970 Porto Alegre, RS, Brazil
| |
Collapse
|
7
|
Modeling Tubercular ESX-1 Secretion Using Mycobacterium marinum. Microbiol Mol Biol Rev 2020; 84:84/4/e00082-19. [DOI: 10.1128/mmbr.00082-19] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Pathogenic mycobacteria cause chronic and acute diseases ranging from human tuberculosis (TB) to nontubercular infections.
Mycobacterium tuberculosis
causes both acute and chronic human tuberculosis. Environmentally acquired nontubercular mycobacteria (NTM) cause chronic disease in humans and animals. Not surprisingly, NTM and
M. tuberculosis
often use shared molecular mechanisms to survive within the host. The ESX-1 system is a specialized secretion system that is essential for virulence and is functionally conserved between
M. tuberculosis
and
Mycobacterium marinum
.
Collapse
|
8
|
Akram MS, Pery N, Butler L, Shafiq MI, Batool N, Rehman MFU, Grahame-Dunn LG, Yetisen AK. Challenges for biosimilars: focus on rheumatoid arthritis. Crit Rev Biotechnol 2020; 41:121-153. [PMID: 33040628 DOI: 10.1080/07388551.2020.1830746] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Healthcare systems worldwide are struggling to find ways to fund the cost of innovative treatments such as gene therapies, regenerative medicine, and monoclonal antibodies (mAbs). As the world's best known mAbs are close to facing patent expirations, the biosimilars market is poised to grow with the hope of bringing prices down for cancer treatment and autoimmune disorders, however, this has yet to be realized. The development costs of biosimilars are significantly higher than their generic equivalents due to therapeutic equivalence trials and higher manufacturing costs. It is imperative that academics and relevant companies understand the costs and stages associated with biologics processing. This article brings these costs to the forefront with a focus on biosimilars being developed for Rheumatoid Arthritis (RA). mAbs have remarkably changed the treatment landscape, establishing their superior efficacy over traditional small chemicals. Five blockbuster TNFα mAbs, considered as first line biologics against RA, are either at the end of their patent life or have already expired and manufacturers are seeking to capture a significant portion of that market. Although in principle, market-share should be available, withstanding that the challenges regarding the compliance and regulations are being resolved, particularly with regards to variation in the glycosylation patterns and challenges associated with manufacturing. Glycan variants can significantly affect the quality attributes requiring characterization throughout production. Successful penetration of biologics can drive down prices and this will be a welcome change for patients and the healthcare providers. Herein we review the biologic TNFα inhibitors, which are on the market, in development, and the challenges being faced by biosimilar manufacturers.
Collapse
Affiliation(s)
- Muhammad Safwan Akram
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK.,National Horizons Centre, Teesside University, Darlington, UK
| | - Neelam Pery
- Institute of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | - Lucy Butler
- School of Health and Life Sciences, Teesside University, Middlesbrough, UK.,National Horizons Centre, Teesside University, Darlington, UK
| | | | - Nayab Batool
- Institute of Biochemistry and Biotechnology, University of the Punjab, Lahore, Pakistan
| | | | | | - Ali K Yetisen
- Department of Chemical Engineering, Imperial College London, London, UK
| |
Collapse
|
9
|
Srinivas C, Odsbu I, Linder M. Risk of common infections among individuals with psoriasis in Sweden: A nationwide cohort study comparing secukinumab to ustekinumab. Pharmacoepidemiol Drug Saf 2020; 29:1562-1569. [PMID: 32975344 PMCID: PMC7756328 DOI: 10.1002/pds.5132] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 07/23/2020] [Accepted: 09/06/2020] [Indexed: 12/31/2022]
Abstract
Purpose To determine risk of respiratory tract infections, urinary tract infections and candidiasis in secukinumab users compared to ustekinumab users among individuals with psoriasis in Sweden. Methods This was a Swedish population‐based register‐linked new‐user cohort study on individuals with psoriasis and psoriasis arthritis treated with secukinumab (2015‐2017) and ustekinumab (2009‐2017). Ever‐never exposure definition was used, that is, each individual's follow‐up time was attributed to the drug they were first exposed to. Risk of severe respiratory and urinary tract infections and candidiasis (diagnosis codes from out‐patient specialist visits and in‐patient hospitalisations) and respiratory and urinary tract infections treated in primary care (proxied by dispensation of antibiotics) was determined by adjusted hazard ratios (HRs) and 95% confidence intervals (CIs) using Cox regression. We also give crude incidence rates and rate ratios. Results In total, 1955 new users of secukinumab (n = 848) and ustekinumab (n = 1107) were identified. There was a slightly increased risk of respiratory and urinary tract infections treated in primary care among secukinumab users compared to ustekinumab users (HR: 1.22, 95% CI: 1.03‐1.43). Non‐significant differences in estimated risk of severe respiratory and urinary tract infections (HR: 0.96, 95% CI: 0.57‐1.61) and candidiasis (HR: 1.80, 95% CI: 0.84‐3.84) treated in the hospital setting were observed. Conclusion We observed a slightly increased risk of respiratory and urinary tract infections treated in primary care among secukinumab users compared to ustekinumab users. Larger studies with longer follow‐up are needed to draw conclusions on relative safety.
Collapse
Affiliation(s)
- Chaitra Srinivas
- Centre for Pharmacoepidemiology, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Ingvild Odsbu
- Centre for Pharmacoepidemiology, Department of Medicine, Karolinska Institutet, Solna, Sweden
| | - Marie Linder
- Centre for Pharmacoepidemiology, Department of Medicine, Karolinska Institutet, Solna, Sweden
| |
Collapse
|
10
|
A retrospective cohort study confirms that prophylactic vaccination is underused in patients on tumor necrosis factor inhibitors. J Am Acad Dermatol 2020; 84:1150-1152. [PMID: 32679277 DOI: 10.1016/j.jaad.2020.07.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2020] [Revised: 07/01/2020] [Accepted: 07/08/2020] [Indexed: 11/21/2022]
|
11
|
Sarkar S, Panda S, Kim B, Raychaudhuri SK, Ghosh A, Raychaudhuri SP. Risk of tuberculosis with anti-tumor necrosis factor-alpha therapy in patients with psoriasis and psoriatic arthritis in Indian population. Indian J Dermatol Venereol Leprol 2020; 86:1-7. [PMID: 31719235 DOI: 10.4103/ijdvl.ijdvl_791_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Anti-tumor necrosis factor-alpha (TNF-α) immunotherapy has revolutionized the treatment of inflammatory diseases, such as psoriasis and psoriatic arthritis. However, a major concern is that patients receiving this therapy have an increased risk of infection, particularly of reactivation of latent tuberculosis (TB). There were an estimated 10.4 million new cases of tuberculosis in 2016, worldwide, and India has one of the largest TB case burden with an estimated incidence of 2.79 million cases of TB in the same year. Anti-TNF agents like etanercept and infliximab are available in India approved for psoriasis and psoriatic arthritis. But long-term use of these agents possesses a risk of reactivation of latent TB. In this review article, we assessed the risk of TB with anti-TNF therapy especially in patients with psoriasis and psoriatic arthritis in India. At the end of the article, we have also suggested a recommendation for screening of latent tuberculosis and its management, before starting anti-TNF-α therapy.
Collapse
Affiliation(s)
- Soumajyoti Sarkar
- Division of Rheumatology, Allergy and Clinical Immunology, University of California, Davis, CA, USA
| | - Saumya Panda
- Department of Dermatology, KPC Medical College, Kolkata, West Bengal, India
| | - Byungsoo Kim
- Department of Dermatology, School of Medicine, Biomedical Research Institute, Pusan National University Hospital, Busan, Korea; Department of Dermatology, School of Medicine, University of California, Davis, CA, USA
| | | | - Asutosh Ghosh
- Department of Critical Care Medicine, IPGME and R, Kolkata, West Bengal, India
| | - Siba P Raychaudhuri
- Division of Rheumatology, Allergy and Clinical Immunology; Department of Dermatology, School of Medicine, University of California, Davis; VA Medical Center Sacramento, Mather, CA, USA
| |
Collapse
|
12
|
Choi K, Deval N, Vyas A, Moran C, Cha SS, Mertz LE, Pasha SF, Yiannias JA, Blair JE. The Utility of Screening for Coccidioidomycosis in Recipients of Inhibitors of Tumor Necrosis Factor α. Clin Infect Dis 2020; 68:1024-1030. [PMID: 30084971 DOI: 10.1093/cid/ciy620] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 08/01/2018] [Indexed: 11/14/2022] Open
Abstract
BACKGROUND Tumor necrosis factor α inhibitors (TNFi) are commonly used to treat immune-mediated disorders, but they are associated with an increased risk of mycobacterial and fungal infections. We compared the outcomes of TNFi recipients screened for asymptomatic coccidioidomycosis with those of unscreened patients to compare the development of symptomatic coccidioidomycosis and to describe its outcomes for patients with abnormal coccidioidal screenings. METHODS We searched electronic health records from 4 September 2010 through 26 September 2016 for all patients receiving a TNFi for dermatologic, rheumatologic, or gastroenterologic diagnoses, then categorized patients by whether or not they had undergone coccidioidal serologic testing for screening or diagnostic purposes. RESULTS A total of 2793 patients had a TNFi prescribed. Of those, 1951 met the inclusion criteria: 1025/1951 (52.5%) never had coccidioidal screening; 925/1951 (47.4%) had serologic screening either before beginning TNFi therapy or annually, or both after beginning a TNFi. Symptomatic coccidioidomycosis developed in 35/1025 (3.4%) unscreened patients. Of those screened, 861/925 (93.1%) had negative serologic tests, of which 11/861 (1.3%) subsequently developed symptomatic coccidioidomycosis; 36/925 (3.9%) had coccidioidomycosis at screening (7, probable infection; 11, possible infection; 18, asymptomatic seropositive result); and 17 had only positive findings for immunoglobulin M antibodies and did not meet the definition for coccidioidomycosis. The unscreened cohort was more likely to have symptomatic coccidioidomycosis than the screened cohort (35/1025 vs 11/861, P < .01). CONCLUSIONS Screening for asymptomatic coccidioidomycosis within a Coccidioides-endemic area allowed for identifying and managing asymptomatic coccidioidomycosis before patients began TNFi therapy. Less symptomatic infection developed in the screened than the unscreened cohort.
Collapse
Affiliation(s)
- Kristal Choi
- Division of Rheumatology, Loma Linda University, California
| | - Neha Deval
- Division of Infectious Diseases, Mayo Clinic, Scottsdale, Arizona
| | - Anuj Vyas
- Division of Infectious Diseases, Mayo Clinic, Scottsdale, Arizona
| | - Conor Moran
- School of Graduate Medical Education, College of Medicine and Science, Mayo Clinic, Scottsdale, Arizona
| | - Stephen S Cha
- Division of Biomedical Statistics, Mayo Clinic, Scottsdale, Arizona
| | - Lester E Mertz
- Division of Rheumatology, Mayo Clinic, Scottsdale, Arizona
| | - Shabana F Pasha
- Division of Gastroenterology and Hepatology, Mayo Clinic, Scottsdale, Arizona
| | | | - Janis E Blair
- Division of Infectious Diseases, Mayo Clinic, Scottsdale, Arizona
| |
Collapse
|
13
|
Salado IG, Singh AK, Moreno-Cinos C, Sakaine G, Siderius M, Van der Veken P, Matheeussen A, van der Meer T, Sadek P, Gul S, Maes L, Sterk GJ, Leurs R, Brown D, Augustyns K. Lead Optimization of Phthalazinone Phosphodiesterase Inhibitors as Novel Antitrypanosomal Compounds. J Med Chem 2020; 63:3485-3507. [PMID: 32196340 DOI: 10.1021/acs.jmedchem.9b00985] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Human African trypanosomiasis is causing thousands of deaths every year in the rural areas of Africa. In this manuscript we describe the optimization of a family of phtalazinone derivatives. Phosphodiesterases have emerged as attractive molecular targets for a novel treatment for a variety of neglected parasitic diseases. Compound 1 resulted in being a potent TbrPDEB1 inhibitor with interesting activity against T. brucei in a phenotypic screen. Derivative 1 was studied in an acute in vivo mouse disease model but unfortunately showed no efficacy due to low metabolic stability. We report structural modifications to achieve compounds with an improved metabolic stability while maintaining high potency against TbrPDEB1 and T. brucei. Compound 14 presented a good microsomal stability in mouse and human microsomes and provides a good starting point for future efforts.
Collapse
Affiliation(s)
- Irene G Salado
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Abhimanyu K Singh
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, United Kingdom
| | - Carlos Moreno-Cinos
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Guna Sakaine
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Marco Siderius
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Pieter Van der Veken
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - An Matheeussen
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Tiffany van der Meer
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Payman Sadek
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Sheraz Gul
- Fraunhofer-IME SP, Schnackenburgallee 114, Hamburg 22525, Germany
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| | - Geert-Jan Sterk
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - Rob Leurs
- Medicinal Chemistry, Amsterdam Institute of Molecules, Medicines & Systems, Faculty of Science, Vrije Universiteit Amsterdam, 1081 Amsterdam, The Netherlands
| | - David Brown
- School of Biosciences, University of Kent, Canterbury, Kent CT2 7NJ, United Kingdom
| | - Koen Augustyns
- Laboratory of Medicinal Chemistry, University of Antwerp, Universiteitsplein 1, B-2610 Antwerp, Belgium
| |
Collapse
|
14
|
Attenuating Neurogenic Sympathetic Hyperreflexia Robustly Improves Antibacterial Immunity After Chronic Spinal Cord Injury. J Neurosci 2019; 40:478-492. [PMID: 31754014 DOI: 10.1523/jneurosci.2417-19.2019] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2019] [Revised: 11/11/2019] [Accepted: 11/14/2019] [Indexed: 02/06/2023] Open
Abstract
Spinal cord injury (SCI) disrupts critical physiological systems, including the cardiovascular and immune system. Plasticity of spinal circuits below the injury results in abnormal, heightened sympathetic responses, such as extreme, sudden hypertension that hallmarks life-threatening autonomic dysreflexia. Moreover, such sympathetic hyperreflexia detrimentally impacts other effector organs, including the spleen, resulting in spinal cord injury-induced immunodeficiency. Consequently, infection is a leading cause of mortality after SCI. Unfortunately, there are no current treatments that prophylactically limit sympathetic hyperreflexia to prevent subsequent effector organ dysfunction. The cytokine soluble tumor necrosis factor α (sTNFα) is upregulated in the CNS within minutes after SCI and remains elevated. Here, we report that commencing intrathecal administration of XPro1595, an inhibitor of sTNFα, at a clinically feasible, postinjury time point (i.e., 3 d after complete SCI) sufficiently diminishes maladaptive plasticity within the spinal sympathetic reflex circuit. This results in less severe autonomic dysreflexia, a real-time gauge of sympathetic hyperreflexia, for months postinjury. Remarkably, delayed delivery of the sTNFα inhibitor prevents sympathetic hyperreflexia-associated splenic atrophy and loss of leukocytes to dramatically improve the endogenous ability of chronic SCI rats to fight off pneumonia, a common cause of hospitalization after injury. The improved immune function with XPro1595 correlates with less noradrenergic fiber sprouting and normalized norepinephrine levels in the spleen, indicating that heightened, central sTNFα signaling drives peripheral, norepinephrine-mediated organ dysfunction, a novel mechanism of action. Thus, our preclinical study supports intrathecally targeting sTNFα as a viable strategy to broadly attenuate sympathetic dysregulation, thereby improving cardiovascular regulation and immunity long after SCI.SIGNIFICANCE STATEMENT Spinal cord injury (SCI) significantly disrupts immunity, thus increasing susceptibility to infection, a leading cause of morbidity in those living with SCI. Here, we report that commencing intrathecal administration of an inhibitor of the proinflammatory cytokine soluble tumor necrosis factor α days after an injury sufficiently diminishes autonomic dysreflexia, a real time gauge of sympathetic hyperreflexia, to prevent associated splenic atrophy. This dramatically improves the endogenous ability of chronically injured rats to fight off pneumonia, a common cause of hospitalization. This preclinical study could have a significant impact for broadly improving quality of life of SCI individuals.
Collapse
|
15
|
Rusman T, ten Wolde S, Euser SM, van der Ploeg T, van Hall O, van der Horst‐Bruinsma IE. Gender differences in retention rate of tumor necrosis factor alpha inhibitor treatment in ankylosing spondylitis: a retrospective cohort study in daily practice. Int J Rheum Dis 2018; 21:836-842. [PMID: 29611349 PMCID: PMC5901415 DOI: 10.1111/1756-185x.13271] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
AIM To assess gender differences in ankylosing spondylitis (AS) patients in relation to tumor necrosis factor alpha inhibitor (TNFi) drug survival and occurrence of adverse events in daily practice in a large peripheral hospital. METHOD Retrospective data were collected from AS patients treated with etanercept, infliximab and adalimumab between January 2004 and January 2014. Kaplan-Meier survival curves were conducted to describe the drug survival and occurrence of adverse events in time. RESULTS Overall, 122 AS patients (60.7% male) were included over a 10-year time period, with a mean treatment period of 51 months (1-127 months). In total, 21 (17.2%) patients stopped the TNFi, mainly due to inefficacy (52.4%). Female patients showed a significant shorter treatment period compared to males (33.4 vs. 44.9 months). In addition, female patients switched more between TNFi compared to males (26.9% vs. 16.3%) and had a significantly higher risk at developing infections compared to male patients (26% vs.19%). CONCLUSION Females stayed on the same TNFi for a significantly shorter period compared to males (33.4 vs. 44.9 months) and the most important reason to stop or switch the drug was inefficacy. Moreover, females seemed to be more prone to infections during TNFi treatment than males.
Collapse
Affiliation(s)
- Tamara Rusman
- Amsterdam Rheumatology and Immunology CenterDepartment of RheumatologyVU University Medical CenterAmsterdamThe Netherlands
| | | | - Sjoerd M. Euser
- Regional Laboratory of Public HealthKennemerland HaarlemHaarlemThe Netherlands
- Spaarne Gasthuis AcademieHaarlemThe Netherlands
| | - Tjeerd van der Ploeg
- Spaarne Medical Centre AlkmaarHaarlemThe Netherlands
- Spaarne Gasthuis AcademieHaarlemThe Netherlands
| | | | | |
Collapse
|
16
|
Kyriakidis I, Tragiannidis A, Zündorf I, Groll AH. Invasive fungal infections in paediatric patients treated with macromolecular immunomodulators other than tumour necrosis alpha inhibitors. Mycoses 2017; 60:493-507. [DOI: 10.1111/myc.12621] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2016] [Revised: 02/06/2017] [Accepted: 03/07/2017] [Indexed: 12/17/2022]
Affiliation(s)
- Ioannis Kyriakidis
- 2nd Department of Pediatrics; Aristotle University of Thessaloniki; AHEPA University General Hospital; Thessaloniki Greece
| | - Athanasios Tragiannidis
- 2nd Department of Pediatrics; Aristotle University of Thessaloniki; AHEPA University General Hospital; Thessaloniki Greece
| | - Ilse Zündorf
- Institute of Pharmaceutical Biology; Goethe-University of Frankfurt; Frankfurt am Main Germany
| | - Andreas H. Groll
- Infectious Disease Research Program; Center for Bone Marrow Transplantation and Department of Pediatric Hematology/Oncology; University Childrens Hospital; Muenster Germany
| |
Collapse
|
17
|
Tragiannidis A, Kyriakidis I, Zündorf I, Groll AH. Invasive fungal infections in pediatric patients treated with tumor necrosis alpha (TNF-α) inhibitors. Mycoses 2016; 60:222-229. [DOI: 10.1111/myc.12576] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Revised: 09/17/2016] [Accepted: 09/17/2016] [Indexed: 01/09/2023]
Affiliation(s)
- Athanasios Tragiannidis
- Second Department of Pediatrics; AHEPA University General Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Ioannis Kyriakidis
- Second Department of Pediatrics; AHEPA University General Hospital; Aristotle University of Thessaloniki; Thessaloniki Greece
| | - Ilse Zündorf
- Institute of Pharmaceutical Biology; Goethe-University of Frankfurt; Frankfurt am Main Germany
| | - Andreas H. Groll
- Department of Pediatric Hematology and Oncology; Center for Bone Marrow Transplantation; Infectious Disease Research Program; University Childrens Hospital; Muenster Germany
| |
Collapse
|
18
|
NASPGHAN Clinical Report: Surveillance, Diagnosis, and Prevention of Infectious Diseases in Pediatric Patients With Inflammatory Bowel Disease Receiving Tumor Necrosis Factor-α Inhibitors. J Pediatr Gastroenterol Nutr 2016; 63:130-55. [PMID: 27027903 DOI: 10.1097/mpg.0000000000001188] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Children and adolescents with inflammatory bowel disease (IBD) receiving therapy with tumor necrosis factor α inhibitors (anti-TNFα) pose a unique challenge to health care providers in regard to the associated risk of infection. Published experience in adult populations with distinct autoinflammatory and autoimmune diseases treated with anti-TNFα therapies demonstrates an increased risk of serious infections with intracellular bacteria, mycobacteria, fungi, and some viruses; however, there is a paucity of robust pediatric data. With a rising incidence of pediatric IBD and increasing use of biologic therapies, heightened knowledge and awareness of infections in this population is important for primary care pediatricians, pediatric gastroenterologists, and infectious disease (ID) physicians. This clinical report is the result of a consensus review performed by pediatric ID and gastroenterology physicians detailing relevant published literature regarding infections in pediatric patients with IBD receiving anti-TNFα therapies. The objective of this document is to provide comprehensive information for prevention, surveillance, and diagnosis of infections based on current knowledge, until additional pediatric data are available to inform evidence-based recommendations.
Collapse
|
19
|
Depletion of Alveolar Macrophages Does Not Prevent Hantavirus Disease Pathogenesis in Golden Syrian Hamsters. J Virol 2016; 90:6200-6215. [PMID: 27099308 PMCID: PMC4936146 DOI: 10.1128/jvi.00304-16] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2016] [Accepted: 04/11/2016] [Indexed: 12/12/2022] Open
Abstract
UNLABELLED Andes virus (ANDV) is associated with a lethal vascular leak syndrome in humans termed hantavirus pulmonary syndrome (HPS). The mechanism for the massive vascular leakage associated with HPS is poorly understood; however, dysregulation of components of the immune response is often suggested as a possible cause. Alveolar macrophages are found in the alveoli of the lung and represent the first line of defense to many airborne pathogens. To determine whether alveolar macrophages play a role in HPS pathogenesis, alveolar macrophages were depleted in an adult rodent model of HPS that closely resembles human HPS. Syrian hamsters were treated, intratracheally, with clodronate-encapsulated liposomes or control liposomes and were then challenged with ANDV. Treatment with clodronate-encapsulated liposomes resulted in significant reduction in alveolar macrophages, but depletion did not prevent pathogenesis or prolong disease. Depletion also did not significantly reduce the amount of virus in the lung of ANDV-infected hamsters but altered neutrophil recruitment, MIP-1α and MIP-2 chemokine expression, and vascular endothelial growth factor (VEGF) levels in hamster bronchoalveolar lavage (BAL) fluid early after intranasal challenge. These data demonstrate that alveolar macrophages may play a limited protective role early after exposure to aerosolized ANDV but do not directly contribute to hantavirus disease pathogenesis in the hamster model of HPS. IMPORTANCE Hantaviruses continue to cause disease worldwide for which there are no FDA-licensed vaccines, effective postexposure prophylactics, or therapeutics. Much of this can be attributed to a poor understanding of the mechanism of hantavirus disease pathogenesis. Hantavirus disease has long been considered an immune-mediated disease; however, by directly manipulating the Syrian hamster model, we continue to eliminate individual immune cell types. As the most numerous immune cells present in the respiratory tract, alveolar macrophages are poised to defend against hantavirus infection, but those antiviral responses may also contribute to hantavirus disease. Here, we demonstrate that, like in our prior T and B cell studies, alveolar macrophages neither prevent hantavirus infection nor cause hantavirus disease. While these studies reflect pathogenesis in the hamster model, they should help us rule out specific cell types and prompt us to consider other potential mechanisms of disease in an effort to improve the outcome of human HPS.
Collapse
|
20
|
Veerman J, van den Bergh T, Orrling KM, Jansen C, Cos P, Maes L, Chatelain E, Ioset JR, Edink EE, Tenor H, Seebeck T, de Esch I, Leurs R, Sterk GJ. Synthesis and evaluation of analogs of the phenylpyridazinone NPD-001 as potent trypanosomal TbrPDEB1 phosphodiesterase inhibitors and in vitro trypanocidals. Bioorg Med Chem 2016; 24:1573-81. [PMID: 26935942 DOI: 10.1016/j.bmc.2016.02.032] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/21/2016] [Accepted: 02/24/2016] [Indexed: 12/16/2022]
Abstract
Trypanosomal phosphodiesterases B1 and B2 (TbrPDEB1 and TbrPDEB2) play an important role in the life cycle of Trypanosoma brucei, the causative parasite of human African trypanosomiasis (HAT), also known as African sleeping sickness. Knock down of both enzymes leads to cell cycle arrest and is lethal to the parasite. Recently, we reported the phenylpyridazinone, NPD-001, with low nanomolar IC50 values on both TbrPDEB1 (IC50: 4nM) and TbrPDEB2 (IC50: 3nM) (J. Infect. Dis.2012, 206, 229). In this study, we now report on the first structure activity relationships of a series of phenylpyridazinone analogs as TbrPDEB1 inhibitors. A selection of compounds was also shown to be anti-parasitic. Importantly, a good correlation between TbrPDEB1 IC50 and EC50 against the whole parasite was observed. Preliminary analysis of the SAR of selected compounds on TbrPDEB1 and human PDEs shows large differences which shows the potential for obtaining parasite selective PDE inhibitors. The results of these studies support the pharmacological validation of the Trypanosome PDEB family as novel therapeutic approach for HAT and provide as well valuable information for the design of potent TbrPDEB1 inhibitors that could be used for the treatment of this disease.
Collapse
Affiliation(s)
- Johan Veerman
- Mercachem, PO Box 6747, 6503 GE Nijmegen, The Netherlands
| | | | - Kristina M Orrling
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Chimed Jansen
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Paul Cos
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Groenenborgerlaan 171, 2020 Wilrijk, Belgium
| | - Louis Maes
- Laboratory for Microbiology, Parasitology and Hygiene (LMPH), University of Antwerp, Groenenborgerlaan 171, 2020 Wilrijk, Belgium
| | - Eric Chatelain
- DNDi (Drugs for Neglected Diseases initiative), 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Jean-Robert Ioset
- DNDi (Drugs for Neglected Diseases initiative), 15 Chemin Louis Dunant, 1202 Geneva, Switzerland
| | - Ewald E Edink
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Hermann Tenor
- Takeda, Takeda Pharmaceuticals International GmbH, Thurgauerstrasse 130, 8152 Glattpark-Opfikon, Zurich, Switzerland
| | - Thomas Seebeck
- Institute of Cell Biology, University of Bern, Baltzerstrasse 4, 3012 Bern, Switzerland
| | - Iwan de Esch
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Rob Leurs
- Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands
| | - Geert Jan Sterk
- Mercachem, PO Box 6747, 6503 GE Nijmegen, The Netherlands; Division of Medicinal Chemistry, Faculty of Sciences, Amsterdam Institute of Molecules, Medicines & Systems (AIMMS), Vrije Universiteit Amsterdam, De Boelelaan 1083, 1081 HV Amsterdam, The Netherlands.
| |
Collapse
|
21
|
Sehgal VN, Pandhi D, Khurana A. Biologics in dermatology: adverse effects. Int J Dermatol 2015; 54:1442-60. [PMID: 26147909 DOI: 10.1111/ijd.12802] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2013] [Revised: 01/13/2014] [Accepted: 06/25/2014] [Indexed: 12/13/2022]
Abstract
Biologics are a group of drugs that precisely affect certain specific steps in the immune response and are an extremely useful group when used in an appropriate setting. However, their use can often be a double-edged sword. Careful patient selection and thorough knowledge of adverse effects is a key to their successful use in various disorders. The initial enthusiasm has gradually given way to a more cautious approach wherein a balance is sought between clinical usefulness and expected side effects. The adverse effects of the biologics most commonly used in dermatology have been carefully listed for ready reference. The plausible causes of the adverse reactions are succinctly outlined along with their incriminating factor(s). Besides, in brief, the attention has been focused on their management. The content should provide an essential didactic content for educating the practitioner.
Collapse
Affiliation(s)
- Virendra N Sehgal
- Dermato-Venereology (Skin/VD) Center, Sehgal Nursing Home, Delhi, India
| | - Deepika Pandhi
- Department of Dermatology and STD, University College of Medical Sciences, and Associated Guru Teg Bahadur Hospital, Shahdara, Delhi, India
| | - Ananta Khurana
- Department of Dermatology and STD, Dr RML hospital and PGIMER, New Delhi, India
| |
Collapse
|
22
|
Menon R, David BG. Itolizumab - a humanized anti-CD6 monoclonal antibody with a better side effects profile for the treatment of psoriasis. Clin Cosmet Investig Dermatol 2015; 8:215-22. [PMID: 25945063 PMCID: PMC4407739 DOI: 10.2147/ccid.s47784] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Management of psoriasis is a challenge to the treating physician. The chronic inflammatory state of psoriasis with exacerbations and remissions necessitate “on-and-off” treatment schedules. The safety profiles of drugs and tolerability issues for patients are important factors to be considered during treatment. Various biological agents targeting T-cells and the inflammatory cytokines are available for systemic treatment of psoriasis. However, major causes of concern while using these drugs are risk of susceptibility to infection and development of anti-drug antibodies, which will affect the pharmacokinetic properties, efficacy, and safety profile of the drug. Itolizumab, a humanized anti-CD6 monoclonal antibody, is a new molecule that acts by immunomodulating the CD6 molecule. CD6 is a co-stimulatory molecule required for optimal T-cell stimulation by the antigen-presenting cells. This step is crucial in T-cell proliferation to form Th1 and Th17 cells, which play a major role in the pathogenesis of psoriasis. This article deals with the properties of Itolizumab and its role in the treatment of psoriasis. Based on the available published data, Itolizumab seems to have a better adverse effects profile and at the same time comparatively less efficacy when compared to other biological agents available for treating psoriasis. Larger studies with longer duration are required to clearly depict the long-term side effects profile.
Collapse
Affiliation(s)
- Roshni Menon
- Department of Dermatology, Venereology and Leprosy, Sri Venkateshwaraa Medical College Hospital and Research Centre, Ariyur, Pondicherry, India
| | - Brinda G David
- Department of Dermatology, Venereology and Leprosy, Sri Venkateshwaraa Medical College Hospital and Research Centre, Ariyur, Pondicherry, India
| |
Collapse
|
23
|
Willrich MAV, Murray DL, Snyder MR. Tumor necrosis factor inhibitors: clinical utility in autoimmune diseases. Transl Res 2015; 165:270-82. [PMID: 25305470 DOI: 10.1016/j.trsl.2014.09.006] [Citation(s) in RCA: 100] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2014] [Revised: 09/09/2014] [Accepted: 09/10/2014] [Indexed: 12/17/2022]
Abstract
Tumor necrosis factor (TNF) production is amplified in several autoimmune disorders. In the 1990s, it became a validated therapeutic target used for the treatment of conditions such as rheumatoid arthritis and inflammatory bowel disease. Biologic drugs targeting TNF include engineered monoclonal antibodies and fusion proteins. Currently, there are 5 Food and Drug Administration-approved TNF inhibitors (infliximab, etanercept, adalimumab, certolizumab, and golimumab), representing close to $20 billion in sales. Clinical trials remain open to test their efficacy and safety compared with one another, as well as to measure clinical outcomes in different conditions and patient populations. The industry is also eager to develop biotherapeutics that are similar but cheaper than the currently existing biologics or are safer with higher efficacy; these are the so-called "biosimilars." Clinical utility of TNF inhibitors and indications of mono- or combined therapy with immunomodulators are reviewed here. Pharmacokinetics of the TNF inhibitors is affected by routes of administration, clearance mechanisms of immunoglobulins, and immunogenicity. Finally, strategies for management of treatment efficacy and increasing evidence for monitoring of serum concentration of TNF inhibitors are discussed, assessing for the presence of the antidrug antibodies and the different analytical methods available for laboratory testing. As clinical applications of the TNF inhibitors expand, and other classes join the revolution in the treatment of chronic inflammatory disorders, therapeutic drug monitoring of biologics will become increasingly important, with the potential to dramatically improve patient care and management.
Collapse
Affiliation(s)
- Maria A V Willrich
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - David L Murray
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn
| | - Melissa R Snyder
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minn.
| |
Collapse
|
24
|
Pushalkar S, Li X, Kurago Z, Ramanathapuram LV, Matsumura S, Fleisher KE, Glickman R, Yan W, Li Y, Saxena D. Oral microbiota and host innate immune response in bisphosphonate-related osteonecrosis of the jaw. Int J Oral Sci 2014; 6:219-26. [PMID: 25105817 PMCID: PMC5153588 DOI: 10.1038/ijos.2014.46] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2014] [Indexed: 12/31/2022] Open
Abstract
Bacterial biofilms have emerged as potential critical triggers in the pathogenesis of bisphosphonate (BP)-related osteonecrosis of the jaw (ONJ) or BRONJ. BRONJ lesions have shown to be heavily colonized by oral bacteria, most of these difficult to cultivate and presents many clinical challenges. The purpose of this study was to characterize the bacterial diversity in BRONJ lesions and to determine host immune response. We examined tissue specimens from three cohorts (n=30); patients with periodontal disease without a history of BP therapy (Control, n=10), patients with periodontal disease having history of BP therapy but without ONJ (BP, n=5) and patients with BRONJ (BRONJ, n=15). Denaturing gradient gel electrophoresis of polymerase chain reaction (PCR)-amplified 16S rRNA gene fragments revealed less bacterial diversity in BRONJ than BP and Control cohorts. Sequence analysis detected six phyla with predominant affiliation to Firmicutes in BRONJ (71.6%), BP (70.3%) and Control (59.1%). Significant differences (P<0.05) in genera were observed, between Control/BP, Control/BRONJ and BP/BRONJ cohorts. Enzyme-linked immunosorbent assay (ELISA) results indicated that the levels of myeloperoxidase were significantly lower, whereas interleukin-6 and tumor necrosis factor-alpha levels were moderately elevated in BRONJ patients as compared to Controls. PCR array showed significant changes in BRONJ patients with downregulation of host genes, such as nucleotide-binding oligomerization domain containing protein 2, and cathepsin G, the key modulators for antibacterial response and upregulation of secretory leukocyte protease inhibitor, proteinase 3 and conserved helix-loop-helix ubiquitous kinase. The results suggest that colonization of unique bacterial communities coupled with deficient innate immune response is likely to impact the pathogenesis of ONJ.
Collapse
Affiliation(s)
- Smruti Pushalkar
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, USA
| | - Xin Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, USA
| | - Zoya Kurago
- Department of Oral and Maxillofacial Pathology, Radiology and Medicine, New York University College of Dentistry, New York, USA
| | - Lalitha V Ramanathapuram
- Department of Oral and Maxillofacial Pathology, Radiology and Medicine, New York University College of Dentistry, New York, USA
| | - Satoko Matsumura
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, USA
| | - Kenneth E Fleisher
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, USA
| | - Robert Glickman
- Department of Oral and Maxillofacial Surgery, New York University College of Dentistry, New York, USA
| | - Wenbo Yan
- Department of Biology, Nyack College, New York, USA
| | - Yihong Li
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, USA
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, USA
| |
Collapse
|
25
|
Prieto-Pérez R, Cabaleiro T, Daudén E, Ochoa D, Román M, Abad-Santos F. Pharmacogenetics of topical and systemic treatment of psoriasis. Pharmacogenomics 2014; 14:1623-34. [PMID: 24088133 DOI: 10.2217/pgs.13.163] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Psoriasis is a chronic inflammatory skin disease. The cause of psoriasis is unknown, although genetics may play a key role in its development. Treatment of the disease varies with severity. Topical drugs, such as corticosteroids, coal tar, retinoids and vitamin D analogs, are commonly used to treat mild psoriasis. Phototherapy and systemic drugs, such as calcineurin inhibitors, methotrexate, acitretin and biological drugs, are usually used to treat moderate-to-severe psoriasis. Not all patients respond well to treatment, and some can develop severe adverse effects. Interindividual differences in several genes may explain this variation in response to treatment. Pharmacogenetics and pharmacogenomics can facilitate more personalized medicine and prevent the adverse effects associated with treatment.
Collapse
Affiliation(s)
- Rocío Prieto-Pérez
- Service of Clinical Pharmacology, Hospital Universitario de la Princesa, Instituto Teófilo Hernando, Universidad Autónoma de Madrid, Instituto de Investigación Sanitaria Princesa (IP), Diego de León 62, 28006 Madrid, Spain
| | | | | | | | | | | |
Collapse
|
26
|
Solovan C, Chiticariu E. Psoriasis, anti-tumor necrosis factor therapy, and tuberculosis: report of three challenging cases and literature review. Infect Dis Ther 2013; 2:59-73. [PMID: 25135824 PMCID: PMC4108098 DOI: 10.1007/s40121-013-0003-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2012] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION The era of biologic therapies has provided new options for the treatment of chronic plaque psoriasis. However, safety concerns have led to intensive screening and monitoring of patients receiving anti-tumor necrosis factor alpha (anti-TNF-alpha) agents. METHODS The authors describe the cases of three patients with moderate to severe psoriasis treated with anti-TNF agents, with challenging diagnostic and treatment aspects regarding tuberculosis (TB) infection, a serious adverse event associated with this type of treatment. The cases are discussed in the context of a comprehensive literature review describing the risk of TB associated with the use of TNF inhibitors. A critical review of the clinical trials that have tested the safety of these agents is also presented. RESULTS One patient, who tested negatively for latent TB infection (LTBI) during screening, developed active TB under adalimumab therapy. For two other patients the diagnosis and management of LTBI in relation to anti-TNF therapy represented a challenge. Although clinical trials involving the use of anti-TNF therapy for psoriasis haven't demonstrated a high TB incidence, active TB is continuously reported in association with this treatment. CONCLUSIONS Findings from clinical practice and the scientific literature indicate that anti-TNF therapies are associated with an increased risk of TB, and close monitoring of patients is needed.
Collapse
Affiliation(s)
- Caius Solovan
- Department of Dermatology, University of Medicine and Pharmacy “Victor Babes” Timisoara, Marasesti 5, 300077 Timisoara, Romania
| | - Elena Chiticariu
- Department of Dermatology, University of Medicine and Pharmacy “Victor Babes” Timisoara, Marasesti 5, 300077 Timisoara, Romania
| |
Collapse
|
27
|
Safety of tumor necrosis factor inhibitors use for rheumatoid arthritis and ankylosing spondylitis in Africa, the Middle East, and Asia: focus on severe infections and tuberculosis. Clin Rheumatol 2012; 32:293-300. [PMID: 23242389 DOI: 10.1007/s10067-012-2137-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2012] [Revised: 08/15/2012] [Accepted: 10/19/2012] [Indexed: 01/09/2023]
Abstract
Multiple studies of patients in Western countries with rheumatoid arthritis (RA) and ankylosing spondylitis (AS) have indicated increased risk for active tuberculosis (TB) and other infections among these individuals. It has also been consistently reported that patients receiving tumor necrosis factor (TNF) inhibitors for these conditions have higher rates of active TB and other infections than RA or AS patients not receiving these medications. These issues have been studied less extensively in the Asia and Africa-Middle East regions, and information from these regions is important because of higher rates of TB in the general population. This paper reviews studies of RA and AS patients from Asia, Africa, and the Middle East who received TNF inhibitors. A literature search was conducted using http://www.ncbi.nlm.nih.gov/pubmed to collect and report these data. The years included in the PubMed literature search ranged from January 2000 to October 2011. Additionally, information from the China Hospital Knowledge Database was used to report data from Chinese patients with RA and AS treated with TNF inhibitors. Results from these studies indicate that the risk for active TB and other infections in AS and RA patients from Asia, Africa, and the Middle East are increased in patients receiving TNF inhibitors and that the risk is higher among those treated with monoclonal antibodies versus soluble TNF receptor.
Collapse
|
28
|
|
29
|
Polachek A, Caspi D, Elkayam O. The perioperative use of biologic agents in patients with rheumatoid arthritis. Autoimmun Rev 2012; 12:164-8. [DOI: 10.1016/j.autrev.2012.04.001] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2012] [Accepted: 04/04/2012] [Indexed: 12/20/2022]
|
30
|
Sánchez-Moya A, García-Doval I, Carretero G, Sánchez-Carazo J, Ferrandiz C, Herrera Ceballos E, Alsina M, Ferrán M, López-Estebaranz JL, Gómez-García F, De la Cueva Dobao P, Carrascosa JM, Vanaclocha F, Belinchón I, Peral F, Dauden E. Latent tuberculosis infection and active tuberculosis in patients with psoriasis: a study on the incidence of tuberculosis and the prevalence of latent tuberculosis disease in patients with moderate-severe psoriasis in Spain. BIOBADADERM registry. J Eur Acad Dermatol Venereol 2012; 27:1366-74. [DOI: 10.1111/jdv.12011] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
31
|
Piro JR, Benjamin DI, Duerr JM, Pi Y, Gonzales C, Wood KM, Schwartz JW, Nomura DK, Samad TA. A dysregulated endocannabinoid-eicosanoid network supports pathogenesis in a mouse model of Alzheimer's disease. Cell Rep 2012; 1:617-23. [PMID: 22813736 DOI: 10.1016/j.celrep.2012.05.001] [Citation(s) in RCA: 169] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/07/2012] [Accepted: 05/02/2012] [Indexed: 12/29/2022] Open
Abstract
Although inflammation in the brain is meant as a defense mechanism against neurotoxic stimuli, increasing evidence suggests that uncontrolled, chronic, and persistent inflammation contributes to neurodegeneration. Most neurodegenerative diseases have now been associated with chronic inflammation, including Alzheimer's disease (AD). Whether anti-inflammatory approaches can be used to treat AD, however, is a major unanswered question. We recently demonstrated that monoacylglycerol lipase (MAGL) hydrolyzes endocannabinoids to generate the primary arachidonic acid pool for neuroinflammatory prostaglandins. In this study, we show that genetic inactivation of MAGL attenuates neuroinflammation and lowers amyloid β levels and plaques in an AD mouse model. We also find that pharmacological blockade of MAGL recapitulates the cytokine-lowering effects through reduced prostaglandin production, rather than enhanced endocannabinoid signaling. Our findings thus reveal a role of MAGL in modulating neuroinflammation and amyloidosis in AD etiology and put forth MAGL inhibitors as a potential next-generation strategy for combating AD.
Collapse
Affiliation(s)
- Justin R Piro
- Neuroscience Research Unit, Pfizer Global Research and Development, Groton, CT 06340, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Raychaudhuri SP. Comorbidities of psoriatic arthritis -- metabolic syndrome and prevention: a report from the GRAPPA 2010 annual meeting. J Rheumatol 2012; 39:437-40. [PMID: 22298276 DOI: 10.3899/jrheum.111244] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Psoriatic arthritis (PsA) is associated with serious comorbidities such as increased cardiovascular risk, hypertension, depression, and reduced quality of life. Patients with psoriasis have been observed to have an increased incidence of metabolic syndrome compared with the general population; recently, this has also been observed in patients with PsA. This review focuses on the comorbidities associated with PsA, with an emphasis on risks of coronary artery disease and metabolic syndrome. We also discuss the development of a comprehensive approach for the management of comorbidities of PsA. The review summarizes a presentation at the 2010 annual meeting of GRAPPA (Group for Research and Assessment of Psoriasis and Psoriatic Arthritis).
Collapse
Affiliation(s)
- Siba P Raychaudhuri
- Veterans Affairs Sacramento Medical Center, University of California Davis, School of Medicine, Division of Rheumatology, Allergy and Clinical Immunology, Davis, California, USA.
| |
Collapse
|
33
|
Dentan C, Mazet R, Gilson M, Marchou-Lopez S, Gaudin P. Rheumatoid arthritis, alveolar echinococcosis, and rituximab: A case report. Joint Bone Spine 2012; 79:325-7. [DOI: 10.1016/j.jbspin.2011.10.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/03/2011] [Indexed: 11/24/2022]
|
34
|
Knight C, Mauskopf J, Ekelund M, Singh A, Yang S, Boggs R. Cost-effectiveness of treatment with etanercept for psoriasis in Sweden. THE EUROPEAN JOURNAL OF HEALTH ECONOMICS : HEPAC : HEALTH ECONOMICS IN PREVENTION AND CARE 2012; 13:145-56. [PMID: 21380772 DOI: 10.1007/s10198-010-0293-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2009] [Accepted: 12/22/2010] [Indexed: 05/25/2023]
Abstract
OBJECTIVE To estimate the cost-effectiveness, from a Swedish societal perspective, of intermittent use of etanercept (Enbrel) with interruptions of use after 24 weeks compared to continuous use of adalimumab (Humira) as well as non-systemic standard of care in patients with moderate to severe psoriasis. METHODS A Markov decision-tree model was constructed from clinical trials results. Patients starting etanercept, adalimumab, or non-systemic therapy moved through the model's 10-years horizon. Model input parameters included clinical response rates. Outcome measures included direct and indirect costs and quality-adjusted life-years (QALYs). RESULTS The incremental total (direct and indirect) costs per QALY were 1,559,939 kr (<euro>165,354) for adalimumab 40 mg every other week, compared with intermittent once-weekly Enbrel 50 mg, and 93,629 kr (<euro>9,925) for once-weekly intermittent etanercept 50 mg compared with non-systemic standard of care. CONCLUSIONS This analysis showed that, with a 470,000 kr (<euro>50,000) per QALY willingness-to-pay threshold, once-weekly etanercept 50 mg, used intermittently, is a cost-effective treatment for moderate to severe psoriasis compared with adalimumab and non-systemic standard of care.
Collapse
Affiliation(s)
- Christopher Knight
- RTI Health Solutions, Velocity House Business and Conference Centre, Sheffield, UK.
| | | | | | | | | | | |
Collapse
|
35
|
Hepatitis B virus reactivation in rheumatoid arthritis and ankylosing spondylitis patients treated with anti-TNFα agents: a retrospective analysis of 49 cases. Clin Rheumatol 2012; 31:931-6. [PMID: 22349880 DOI: 10.1007/s10067-012-1960-1] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2011] [Revised: 01/26/2012] [Accepted: 02/03/2012] [Indexed: 12/16/2022]
Abstract
Clinical guidelines regarding anti-viral prophylaxis for HBV surface antigen (HBsAg) carriers starting anti-TNFα agents are not yet fully established, even in endemic regions of HBV infection. We retrospectively collected the clinical data of 52 HBsAg carriers with rheumatoid arthritis (RA) or ankylosing spondylitis (AS) that had been administered anti-TNFα treatment at seven medical centers in South Korea. Periodic data of liver function tests and serum HBV DNA were both utilized to assess HBV reactivation. The YMDD motif mutation of HBV DNA polymerase was tested in lamivudine-treated patients with elevated HBV DNA. Three of the 52 patients were excluded from the analysis. Of the 49 analyzed patients, 20 patients received anti-viral prophylaxis (15 lamivudine, five entecavir) with anti-TNFα treatment. The remaining 29 patients were treated with anti-viral agents if needed at the discretion of the clinician and did not receive prophylaxis. Of the 29 patients who did not receive primary prophylaxis, two (6.9%) developed viral reactivation within a year of anti-TNFα treatment. In the prophylaxis group, one patient developed viral reactivation at week 64 of anti-TNFα therapy attributed to YMDD mutation caused by lamivudine. Patients with HBV reactivation all responded well to anti-viral therapy. In summary, anti-viral prophylaxis helped preventing HBV reactivation in HBsAg carriers with RA or AS starting anti-TNFα, yet mutation in the YMDD motif of HBV DNA polymerase could be detrimental to some patients under long-term lamivudine prophylaxis.
Collapse
|
36
|
Gentile G, Foà R. Viral infections associated with the clinical use of monoclonal antibodies. Clin Microbiol Infect 2011; 17:1769-75. [DOI: 10.1111/j.1469-0691.2011.03680.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
37
|
Mittal M, Raychaudhuri SP. Golimumab and certolizumab: the two new anti-tumor necrosis factor kids on the block. Indian J Dermatol Venereol Leprol 2011; 76:602-8; quiz 609. [PMID: 21079302 DOI: 10.4103/0378-6323.72445] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
Anti-tumor necrosis factor (anti-TNF) agents have revolutionized treatment of psoriasis and many other inflammatory diseases of autoimmune origin. They have considerable advantages over the existing immunomodulators. Anti-TNF agents are designed to target a very specific component of the immune-mediated inflammatory cascades. Thus, they have lower risks of systemic side-effects. In a brief period of 10 years, a growing number of biological therapies are entering the clinical arena while many more biologicals remain on the horizon. With time, the long-term side-effects and efficacies of these individual agents will become clearer and help to determine which ones are the most suitable for long-term care. Golimumab (a human monoclonal anti-TNF-α antibody) and Certolizumab (a PEGylated Fab fragment of humanized monoclonal TNF-α antibody) are the two latest additions to the anti-TNF regimen. Here, we are providing a brief description about these two drugs and their uses.
Collapse
Affiliation(s)
- Mohit Mittal
- Department of Medicine, Division of Rheumatology, VA Medical Center Sacramento and UC Davis School of Medicine, Allergy and Clinical Immunology, 105 35 Hospital Way, Mather, CA 95655, USA
| | | |
Collapse
|
38
|
Lund BM, O'Brien SJ. The occurrence and prevention of foodborne disease in vulnerable people. Foodborne Pathog Dis 2011; 8:961-73. [PMID: 21561383 PMCID: PMC3159107 DOI: 10.1089/fpd.2011.0860] [Citation(s) in RCA: 123] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
In developed countries, such as the United Kingdom and the United States, between 15% and 20% of the population show greater susceptibility than the general population to foodborne disease. This proportion includes people with primary immunodeficiency, patients treated with radiation or with immunosuppressive drugs for cancer and diseases of the immune system, those with acquired immune-deficiency syndrome and diabetics, people suffering from liver or kidney disease or with excessive iron in the blood, pregnant women, infants, and the elderly. Malnutrition and use of antacids, particularly proton-pump inhibitors, also increase susceptibility. We review the occurrence of infection by foodborne pathogens in these groups of people and measures to prevent infection. The nature and use of low microbial diets to reduce the risk of foodborne disease in immunocompromised patients are very variable. Diets for vulnerable people in care should exclude higher-risk foods, and vulnerable people in the community should receive clear advice about food safety, in particular avoidance of higher-risk foods and substitution of safer, nutritious foods.
Collapse
|
39
|
Jin L, Hill KK, Filak H, Mogan J, Knowles H, Zhang B, Perraud AL, Cambier JC, Lenz LL. MPYS is required for IFN response factor 3 activation and type I IFN production in the response of cultured phagocytes to bacterial second messengers cyclic-di-AMP and cyclic-di-GMP. THE JOURNAL OF IMMUNOLOGY 2011; 187:2595-601. [PMID: 21813776 DOI: 10.4049/jimmunol.1100088] [Citation(s) in RCA: 232] [Impact Index Per Article: 16.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Cyclic-di-GMP and cyclic-di-AMP are second messengers produced by bacteria and influence bacterial cell survival, differentiation, colonization, biofilm formation, virulence, and bacteria-host interactions. In this study, we show that in both RAW264.7 macrophage cells and primary bone marrow-derived macrophages, the production of IFN-β and IL-6, but not TNF, in response to cyclic-di-AMP and cyclic-di-GMP requires MPYS (also known as STING, MITA, and TMEM173). Furthermore, expression of MPYS was required for IFN response factor 3 but not NF-κB activation in response to these bacterial metabolites. We also confirm that MPYS is required for type I IFN production by cultured macrophages infected with the intracellular pathogens Listeria monocytogenes and Francisella tularensis. However, during systemic infection with either pathogen, MPYS deficiency did not impact bacterial burdens in infected spleens. Serum IFN-β and IL-6 concentrations in the infected control and MPYS(-/-) mice were also similar at 24 h postinfection, suggesting that these pathogens stimulate MPYS-independent cytokine production during in vivo infection. Our findings indicate that bifurcating MPYS-dependent and -independent pathways mediate sensing of cytosolic bacterial infections.
Collapse
Affiliation(s)
- Lei Jin
- Integrated Department of Immunology, University of Colorado Denver School of Medicine, Denver, CO 80206, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Cacciapaglia F, Navarini L, Menna P, Salvatorelli E, Minotti G, Afeltra A. Cardiovascular safety of anti-TNF-alpha therapies: Facts and unsettled issues. Autoimmun Rev 2011; 10:631-5. [DOI: 10.1016/j.autrev.2011.04.014] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
41
|
Nguyen CT, Raychaudhuri SP. SCEDOSPORIUM INFECTION IN A PATIENT WITH ANTI-TNFα THERAPY. Indian J Dermatol 2011; 56:82-3. [PMID: 21572800 PMCID: PMC3088944 DOI: 10.4103/0019-5154.77560] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Patients on anti-TNFα therapy are at increased risk for rare opportunistic infections. Here we are reporting a case of Scedosporium apiospermum infection in a patient treated with anti-TNF for 5 years. Patients on anti-TNFα need close follow-up and clinicians should be suspicious for atypical infections in these immunocompromised hosts.
Collapse
Affiliation(s)
- Caroline T Nguyen
- Department of Medicine, VA Medical Center Sacramento and UC Davis School of Medicine, Division of Rheumatology, Allergy and Clinical Immunology , Hospital Way, Mather, CA 95655, USA
| | | |
Collapse
|
42
|
Sivamani RK, Correa G, Ono Y, Bowen MP, Raychaudhuri SP, Maverakis E. Biological therapy of psoriasis. Indian J Dermatol 2011; 55:161-70. [PMID: 20606887 PMCID: PMC2887522 DOI: 10.4103/0019-5154.62754] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023] Open
Abstract
The treatment of psoriasis has undergone a revolution with the advent of biologic therapies, including infliximab, etanercept, adalimumab, efalizumab, and alefacept. These medications are designed to target specific components of the immune system and are a major technological advancement over traditional immunosuppressive medications. These usually being well tolerated are being found useful in a growing number of immune-mediated diseases, psoriasis being just one example. The newest biologic, ustekinumab, is directed against the p40 subunit of the IL-12 and IL-23 cytokines. It has provided a new avenue of therapy for an array of T-cell-mediated diseases. Biologics are generally safe; however, there has been concern over the risk of lymphoma with use of these agents. All anti-TNF-α agents have been associated with a variety of serious and “routine” opportunistic infections.
Collapse
Affiliation(s)
- Raja K Sivamani
- Department of Medicine, Santa Clara Valley Medical Center, San Jose, CA 95128
| | | | | | | | | | | |
Collapse
|
43
|
Workshop Summary: Functions of the TNF Family in Infectious Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2011; 691:171-5. [DOI: 10.1007/978-1-4419-6612-4_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
44
|
Abinun M. An overview of infectious complications in children on new biologic response-modifying agents. ACTA ACUST UNITED AC 2010. [DOI: 10.2217/phe.10.57] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The clinical need for better treatments as well as the significant progress in understanding the pathophysiology of inflammation on one hand, and the progressive development of biotechnology on the other, were the driving force for the emergence of new treatments for autoimmune disorders at the beginning of the 21st century, heralding the ‘age of biologic response modifying agents’ or biologics. This new class of drugs, although in use for just over a decade, has revolutionized the treatment of many inflammatory disorders, such as rheumatic, connective tissue disorders, autoimmune and autoinflammatory diseases. They have already made an immense impact on the quality of life of patients experiencing many years of combined immunosuppressive and anti-inflammatory treatments for chronic and often debilitating diseases. As these drugs were developed with the aim of altering specific components in the immune system function and, in particular, the inflammatory response, it is not surprising that infectious complications, including the severe and unusual, are among the serious side effects alongside other features of dysregulated immune system function, such as autoimmunity, lymphoproliferation and malignancy. The aim of this article is to highlight and anticipate further the infectious risks of the most commonly used biologics in children.
Collapse
Affiliation(s)
- Mario Abinun
- Children’s Bone Marrow Transplantation Unit, Newcastle General Hospital, Westgate Road, Newcastle upon Tyne, NE4 6BE, UK
| |
Collapse
|
45
|
Allie N, Keeton R, Court N, Abel B, Fick L, Vasseur V, Vacher R, Olleros ML, Drutskaya MS, Guler R, Nedospasov SA, Garcia I, Ryffel B, Quesniaux VFJ, Jacobs M. Limited role for lymphotoxin α in the host immune response to Mycobacterium tuberculosis. THE JOURNAL OF IMMUNOLOGY 2010; 185:4292-301. [PMID: 20817877 DOI: 10.4049/jimmunol.1000650] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
The contribution of lymphotoxin (LT)α in the host immune response to virulent Mycobacterium tuberculosis and Mycobacterium bovis bacillus Calmette-Guérin infections was investigated. Despite their ability to induce Th1 cytokine, IFN-γ, and IL-12 pulmonary response, "conventional" LTα(-/-) mice succumb rapidly to virulent M. tuberculosis aerosol infection, with uncontrolled bacilli growth, defective granuloma formation, necrosis, and reduced pulmonary inducible NO synthase expression, similar to TNF(-/-) mice. Contributions from developmental lymphoid abnormalities in LTα(-/-) mice were excluded because hematopoietic reconstitution with conventional LTα(-/-) bone marrow conferred enhanced susceptibility to wild-type mice, comparable to conventional LTα(-/-) control mice. However, conventional LTα(-/-) mice produced reduced levels of TNF after M. bovis bacillus Calmette-Guérin infection, and their lack of control of mycobacterial infection could be due to a defective contribution of either LTα or TNF, or both, to the host immune response. To address this point, the response of "neo-free" LTα(-/-) mice with unperturbed intrinsic TNF expression to M. tuberculosis infection was investigated in a direct comparative study with conventional LTα(-/-) mice. Strikingly, although conventional LTα(-/-) mice were highly sensitive, similar to TNF(-/-) mice, neo-free LTα(-/-) mice controlled acute M. tuberculosis infection essentially as wild-type mice. Pulmonary bacterial burden and inflammation was, however, slightly increased in neo-free LTα(-/-) mice 4-5 mo postinfection, but importantly, they did not succumb to infection. Our findings revise the notion that LTα might have a critical role in host defense to acute mycobacterial infection, independent of TNF, but suggest a contribution of LTα in the control of chronic M. tuberculosis infection.
Collapse
Affiliation(s)
- Nasiema Allie
- Division of Immunology, Institute of Infectious Disease and Molecular Medicine, University of Cape Town, Cape Town, South Africa
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zidi I, Bouaziz A, Mnif W, Bartegi A, Al-Hizab FA, Amor NB. Golimumab therapy of rheumatoid arthritis: an overview. Scand J Immunol 2010; 72:75-85. [PMID: 20618765 DOI: 10.1111/j.1365-3083.2010.02423.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Golimumab is a new approved humanized antibody for the treatment of rheumatoid arthritis (RA). This antibody belonging to biologic agents is raised against the pro-inflammatory cytokine tumour necrosis factor-alpha playing an essential role in the initiation of RA. To date, Golimumab administration for patients with RA, as indicated by USA Food and Drug Administration, is subcutaneous combined with methotrexate (MTX). Here, we have reviewed current literature with a focus on characteristics of Golimumab and also have exposed the clinical trials either using MTX or not using MTX. We have also highlighted the incoming clinical trials on Golimumab and have proposed some indications for the future studies based on a setting of clinical data and post-marketing observational studies. These studies will advance rheumatologists' decisions in the beginning of RA therapeutic interventions to insure the best outcomes for patients with RA and to improve their quality of life.
Collapse
Affiliation(s)
- I Zidi
- Laboratory of Biochemistry, Research Unit 02/UR/09-01, Higher Institute of Biotechnology, Monastir, Tunisia.
| | | | | | | | | | | |
Collapse
|
47
|
Chang C, Gershwin ME. Drugs and autoimmunity--a contemporary review and mechanistic approach. J Autoimmun 2009; 34:J266-75. [PMID: 20015613 DOI: 10.1016/j.jaut.2009.11.012] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Drug-induced autoimmunity is an idiosyncratic, non-IgE immune related drug reaction. Interestingly, although many drugs have been reported to induce autoantibodies, only a few have a definitive association with drug-induced autoimmune disease. The prototype disease is drug-induced lupus and the typical drug for drug-induced lupus is minocycline. The production of autoantibodies and the induction of symptoms in drug-induced lupus results from a variety of mechanisms, which can include suppression of central or peripheral tolerance, alteration of gene transcription in T and B cells, abnormal cytokine and/or cytokine receptor balance and function, chromatin structure modification and antigen modification. Multiple mechanisms may apply for different drugs, and understanding the pharmacological actions of these agents helps us decipher the etiology. For example, DNA hypomethylation may occur with hydralazine, which leads to increased transcription, increased LFA-1, the generation of autoreactive T cells and a breakdown in peripheral tolerance. Frequently, more than one pathway may be involved. Interestingly, most patients with newly formed autoantibodies resulting from drugs do not develop clinical disease. Nonetheless, the explosion in the use of biological modifiers has been associated with production of autoantibodies, an observation that illustrates the complex nature of these interactions, in that these agents are frequently used to treat autoimmunity, yet may produce autoimmune diseases themselves.
Collapse
Affiliation(s)
- Christopher Chang
- Division of Rheumatology, Allergy and Clinical Immunology, University of California at Davis School of Medicine, 451 Health Sciences Drive, Suite 6510, Davis, CA 95616, USA.
| | | |
Collapse
|