1
|
Mansilla-Polo M, Morgado-Carrasco D. Biologics Versus JAK Inhibitors. Part I: Cancer Risk. A Narrative Review. Dermatol Ther (Heidelb) 2024; 14:1389-1442. [PMID: 38763966 PMCID: PMC11169156 DOI: 10.1007/s13555-024-01166-4] [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: 03/06/2024] [Accepted: 04/10/2024] [Indexed: 05/21/2024] Open
Abstract
INTRODUCTION Biological drugs (BD) and Janus kinase inhibitors (JAKi) have revolutionized the treatment of diverse dermatoses. However, there are concerns regarding their safety, especially the risk of cancer and opportunistic infections. Here, we discuss the risk of cancer associated with the BD and JAKi used in dermatology. METHODS A narrative review was carried out. All relevant articles evaluating the risk of cancer associated with BD or JAKi and published between January 2010 and February 2024 were selected. RESULTS Multiple large studies have evaluated the association between BD, JAKi and cancer risk. However, there is a lack of prospective, comparative studies. Overall, patients undergoing BD and JAKi present a cutaneous cancer incidence similar to that in the general population. The drugs more strongly associated with non-skin cancer risk were anti-tumor necrosis factor (anti-TNFs) agents and JAKi (especially tofacitinib and oral ruxolitinib). This risk appears to increase with age, the presence of other factors (such as chronic immunosuppression from previous drugs or other comorbidities), and specific diseases such as rheumatoid arthritis (RA) and myelodysplastic syndrome. Conversely, BD such as interleukin (IL)-17 and IL-23 inhibitors may even reduce the risk of some visceral and hematological malignancies. In patients with dermatological conditions such as psoriasis and atopic dermatitis, the risk of malignancies may be lower than in other subgroups, and probably comparable to the general population. CONCLUSIONS The incidence of cancer in patients undergoing BD or JAKi is generally low. This incidence can be higher in elderly patients with RA or myelodysplastic syndrome, and in those undergoing prolonged therapy with tofacitinib or ruxolitinib (oral), or anti-TNF agents.
Collapse
Affiliation(s)
- Miguel Mansilla-Polo
- Department of Dermatology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Instituto de Investigación Sanitaria (IIS) La Fe, Valencia, Spain
- Department of Dermatology, Faculty of Medicine, Universitat de València, Villarroel 170, 08036, Valencia, Spain
| | - Daniel Morgado-Carrasco
- Department of Dermatology, Hospital Clínic de Barcelona, Universitat de Barcelona, Barcelona, Spain.
- Department of Dermatology, Hospital de Figueres, Fundació Alt Empurdà, Figueres, Spain.
| |
Collapse
|
2
|
Chernikov IV, Bachkova IK, Sen’kova AV, Meschaninova MI, Savin IA, Vlassov VV, Zenkova MA, Chernolovskaya EL. Cholesterol-Modified Anti-Il6 siRNA Reduces the Severity of Acute Lung Injury in Mice. Cells 2024; 13:767. [PMID: 38727303 PMCID: PMC11083178 DOI: 10.3390/cells13090767] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Revised: 04/27/2024] [Accepted: 04/29/2024] [Indexed: 05/13/2024] Open
Abstract
Small interfering RNA (siRNA) holds significant therapeutic potential by silencing target genes through RNA interference. Current clinical applications of siRNA have been primarily limited to liver diseases, while achievements in delivery methods are expanding their applications to various organs, including the lungs. Cholesterol-conjugated siRNA emerges as a promising delivery approach due to its low toxicity and high efficiency. This study focuses on developing a cholesterol-conjugated anti-Il6 siRNA and the evaluation of its potency for the potential treatment of inflammatory diseases using the example of acute lung injury (ALI). The biological activities of different Il6-targeted siRNAs containing chemical modifications were evaluated in J774 cells in vitro. The lead cholesterol-conjugated anti-Il6 siRNA after intranasal instillation demonstrated dose-dependent therapeutic effects in a mouse model of ALI induced by lipopolysaccharide (LPS). The treatment significantly reduced Il6 mRNA levels, inflammatory cell infiltration, and the severity of lung inflammation. IL6 silencing by cholesterol-conjugated siRNA proves to be a promising strategy for treating inflammatory diseases, with potential applications beyond the lungs.
Collapse
Affiliation(s)
- Ivan V. Chernikov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Irina K. Bachkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
- Faculty of Natural Sciences, Novosibirsk State University, Pirogova Str., 1, 630090 Novosibirsk, Russia
| | - Aleksandra V. Sen’kova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Mariya I. Meschaninova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Innokenty A. Savin
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Valentin V. Vlassov
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Marina A. Zenkova
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| | - Elena L. Chernolovskaya
- Institute of Chemical Biology and Fundamental Medicine, Siberian Branch of the Russian Academy of Sciences, Acad. Lavrentiev Ave. 8, 630090 Novosibirsk, Russia; (I.V.C.); (I.K.B.); (A.V.S.); (M.I.M.); (I.A.S.); (M.A.Z.)
| |
Collapse
|
3
|
Bardazzi F, Filippi F, Pagliara A, Clarizio G, Loi C, Sacchelli L, La Placa M. Adalimumab-induced optic neuritis: A rare side effect of biological therapy. Int J Dermatol 2024; 63:e79-e80. [PMID: 38228399 DOI: 10.1111/ijd.17033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 11/26/2023] [Accepted: 01/04/2024] [Indexed: 01/18/2024]
Affiliation(s)
- Federico Bardazzi
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
| | - Federica Filippi
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
| | - Andrea Pagliara
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Giacomo Clarizio
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| | - Camilla Loi
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
| | - Lidia Sacchelli
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
| | - Michelangelo La Placa
- Dermatology Unit - IRCCS, Azienda Ospedaliero-Universitaria, Policlinico Sant'Orsola-Malpighi, Bologna, Italy
- Department of Medical and Surgical Sciences, Alma Mater Studiorum, University of Bologna, Bologna, Italy
| |
Collapse
|
4
|
Little R, Kamath BM, Ricciuto A. Liver Disease in Pediatric Inflammatory Bowel Disease. PEDIATRIC INFLAMMATORY BOWEL DISEASE 2023:129-149. [DOI: 10.1007/978-3-031-14744-9_11] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
5
|
Protracted fever after infliximab induction therapy in a patient with ulcerative colitis (clinical case). ACTA BIOMEDICA SCIENTIFICA 2022. [DOI: 10.29413/abs.2022-7.6.5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Inflammatory bowel diseases (ulcerative colitis and Crohn’s disease) tend to increase in frequency and prevalence worldwide and lead to a significant reduction in quality of life with a complicated course. Intravenous administration of corticosteroids in acute severe ulcerative colitis is effective in 60–70 % of cases. Rescue therapy for patients with steroid‐refractory and steroid-dependence attacks is effective for reducing colectomy or colproctectomy and improves long-term treatment outcomes. Despite the proven efficacy of infliximab, limitations to the use of monoclonal antibodies are adverse reactions to the administration of genetically engineered biologic therapy (GEBT), both well-known and studied, and rare.The aim of the study. To analyze a case of an extremely rare and paradoxical incurable hyperergic reaction that occurred after using genetically engineered biological therapy with tumor necrosis factor alpha blockers in a patient suffering from ulcerative colitis.Materials and methods. We carried out the search in the PubMed, Embase, Scopus and Medline databases until November 1, 2022 using the following keywords: inflammatory bowel disease, ulcerative colitis (UC), anti-tumor necrosis factor α (antiTNFα), infliximab, fever, adverse events. In the scientific electronic library eLIBRARY, a search was made for Russian-language publications using similar queries in Russian. We analyzed the patient’s medical records from 03.06.2013 up to the present day; the patient signed an informed consent for the publication and discussion of the presented data.Conclusion. The widespread use of GEBT in the treatment of autoimmune and autoinflammatory diseases leads to the accumulation of both positive and negative experience of clinicians in real clinical practice. The officially described adverse events in the appointment of any drug suggest options for their prevention and treatment. Atypical manifestations or casuistic cases must be systematized for further fundamental pathophysiological studies.
Collapse
|
6
|
Rommasi F, Nasiri MJ, Mirsaeidi M. Immunomodulatory agents for COVID-19 treatment: possible mechanism of action and immunopathology features. Mol Cell Biochem 2022; 477:711-726. [PMID: 35013850 PMCID: PMC8747854 DOI: 10.1007/s11010-021-04325-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Accepted: 12/01/2021] [Indexed: 02/06/2023]
Abstract
The novel coronavirus pandemic has emerged as one of the significant medical-health challenges of the current century. The World Health Organization has named this new virus severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Since the first detection of SARS-CoV-2 in November 2019 in Wuhan, China, physicians, researchers, and others have made it their top priority to find drugs and cures that can effectively treat patients and reduce mortality rates. The symptoms of Coronavirus Disease 2019 (COVID-19) include fever, dry cough, body aches, and anosmia. Various therapeutic compounds have been investigated and applied to mitigate the symptoms in COVID-19 patients and cure the disease. Degenerative virus analyses of the infection incidence and COVID-19 have demonstrated that SARS-CoV-2 penetrates the pulmonary alveoli's endothelial cells through Angiotensin-Converting Enzyme 2 (ACE2) receptors on the membrane, stimulates various signaling pathways and causes excessive secretion of cytokines. The continuous triggering of the innate and acquired immune system, as well as the overproduction of pro-inflammatory factors, cause a severe condition in the COVID-19 patients, which is called "cytokine storm". It can lead to acute respiratory distress syndrome (ARDS) in critical patients. Severe and critical COVID-19 cases demand oxygen therapy and mechanical ventilator support. Various drugs, including immunomodulatory and immunosuppressive agents (e.g., monoclonal antibodies (mAbs) and interleukin antagonists) have been utilized in clinical trials. However, the studies and clinical trials have documented diverging findings, which seem to be due to the differences in these drugs' possible mechanisms of action. These drugs' mechanism of action generally includes suppressing or modulating the immune system, preventing the development of cytokine storm via various signaling pathways, and enhancing the blood vessels' diameter in the lungs. In this review article, multiple medications from different drug families are discussed, and their possible mechanisms of action are also described.
Collapse
Affiliation(s)
- Foad Rommasi
- Faculty of Life Sciences and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Mohammad Javad Nasiri
- Department of Microbiology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mehdi Mirsaeidi
- Department of Pulmonary and Critical Care, Miller School of Medicine, University of Miami, Miami, FL, USA
| |
Collapse
|
7
|
Abubakar SD, Ihim SA, Farshchi A, Maleknia S, Abdullahi H, Sasaki T, Azizi G. The role of TNF-α and anti-TNF-α agents in the immunopathogenesis and management of immune dysregulation in primary immunodeficiency diseases. Immunopharmacol Immunotoxicol 2022; 44:147-156. [DOI: 10.1080/08923973.2021.2023173] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Affiliation(s)
- Sharafudeen Dahiru Abubakar
- Division of Molecular Pathology, Research Institute for Biomedical Sciences, Tokyo University of Science, Tokyo, Japan
- Department of Medical Laboratory Science, College of Medical Science, Ahmadu Bello University, Zaria, Nigeria
| | - Stella Amarachi Ihim
- Department of Molecular and Cellular Pharmacology, University of Shizuoka, Shizuoka, Japan
- Department of Pharmacology and Toxicology, University of Nigeria, Nsukka, Nigeria
| | - Amir Farshchi
- Biopharmaceutical Research Center, AryoGen Pharmed Inc, Alborz University of Medical Sciences, Karaj, Iran
| | - Shayan Maleknia
- Biopharmaceutical Research Center, AryoGen Pharmed Inc, Alborz University of Medical Sciences, Karaj, Iran
| | - Hamisu Abdullahi
- Department of Immunology, School of Medical Laboratory Sciences, Usmanu Danfodiyo University Sokoto, Sokoto, Nigeria
| | - Takanori Sasaki
- Division of Rheumatology, Immunology and Allergy, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Division of Rheumatology, Department of Internal Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Gholamreza Azizi
- Non-communicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
8
|
Glycerol Improves Skin Lesion Development in the Imiquimod Mouse Model of Psoriasis: Experimental Confirmation of Anecdotal Reports from Patients with Psoriasis. Int J Mol Sci 2021; 22:ijms22168749. [PMID: 34445455 PMCID: PMC8395744 DOI: 10.3390/ijms22168749] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/07/2021] [Accepted: 08/12/2021] [Indexed: 12/19/2022] Open
Abstract
Glycerol is used in many skin care products because it improves skin function. Anecdotal reports by patients on the National Psoriasis Foundation website also suggest that glycerol may be helpful for the treatment of psoriasis, although to date no experimental data confirm this idea. Glycerol entry into epidermal keratinocytes is facilitated by aquaglyceroporins like aquaporin-3 (AQP3), and its conversion to phosphatidylglycerol, a lipid messenger that promotes keratinocyte differentiation, requires the lipid-metabolizing enzyme phospholipase-D2 (PLD2). To evaluate whether glycerol inhibits inflammation and psoriasiform lesion development in the imiquimod (IMQ)-induced mouse model of psoriasis, glycerol’s effect on psoriasiform skin lesions was determined in IMQ-treated wild-type and PLD2 knockout mice, with glycerol provided either in drinking water or applied topically. Psoriasis area and severity index, ear thickness and ear biopsy weight, epidermal thickness, and inflammatory markers were quantified. Topical and oral glycerol ameliorated psoriasiform lesion development in wild-type mice. Topical glycerol appeared to act as an emollient to induce beneficial effects, since even in PLD2 knockout mice topical glycerol application improved skin lesions. In contrast, the beneficial effects of oral glycerol required PLD2, with no improvement in psoriasiform lesions observed in PLD2 knockout mice. Our findings suggest that the ability of oral glycerol to improve psoriasiform lesions requires its PLD2-mediated conversion to phosphatidylglycerol, consistent with our previous report that phosphatidylglycerol itself improves psoriasiform lesions in this model. Our data also support anecdotal evidence that glycerol can ameliorate psoriasis symptoms and therefore might be a useful therapy alone or in conjunction with other treatments.
Collapse
|
9
|
Mashayekhi K, Sankian M, Haftcheshmeh SM, Taheri RA, Hassanpour K, Farnoosh G. A cross-linked anti-TNF-α aptamer for neutralization of TNF-α-induced cutaneous Shwartzman phenomenon: A simple and novel approach for improving aptamers' affinity and efficiency. Biotechnol Prog 2021; 37:e3191. [PMID: 34218531 DOI: 10.1002/btpr.3191] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/26/2021] [Accepted: 07/01/2021] [Indexed: 11/08/2022]
Abstract
To increase the efficiency of aptamers to their targets, a simple and novel method has been developed based on aptamer oligomerization. To this purpose, previously anti-human TNF-α aptamer named T1-T4 was trimerized through a trimethyl aconitate core for neutralization of in vitro and in vivo of TNF-α. At first, 54 mer T1-T4 aptamers with 5'-NH2 groups were covalently coupled to three ester residues in the trimethyl aconitate. In vitro activity of novel anti-TNF-α aptamer and its dissociation constant (Kd ) was done using the L929 cell cytotoxicity assay. In vivo anti-TNF-α activity of new oligomerized aptamer was assessed in a mouse model of cutaneous Shwartzman. Anchoring of three T1-T4 aptamers to trimethyl aconitate substituent results in formation of the 162 mer fragment, which was well revealed by gel electrophoresis. In vitro study indicated that the trimerization of T1-T4 aptamer significantly improved its anti-TNF-α activity compared to non-modified aptamers (P < 0.0001) from 40% to 60%. The determination of Kd showed that trimerization could effectively enhance Kd of aptamer from 67 nM to 36 nM. In vivo study showed that trimer aptamer markedly reduced mean scar size from 15.2 ± 1.2 mm to 1.6 ± 0.1 mm (P < 0.0001), which prevent the formation of skin lesions. In vitro and in vivo studies indicate that trimerization of anti-TNF-α aptamer with a novel approach could improve the anti-TNF-α activity and therapeutic efficacy. According to our findings, a new anti-TNF-α aptamer described here could be considered an appropriate therapeutic agent in treating several inflammatory diseases.
Collapse
Affiliation(s)
- Kazem Mashayekhi
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran.,Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mojtaba Sankian
- Immuno-Biochemistry lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Ramezan Ali Taheri
- Nanobiotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Kazem Hassanpour
- Medical School, Sabzevar University of Medical Sciences, Sabzevar, Iran
| | - Gholamreza Farnoosh
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
10
|
Bury MI, Fuller NJ, Clemons TD, Sturm RM, Morrison CD, Lisy‐Snow DC, Nolan BG, Tarczynski C, Ayello EMT, Boyce A, Muckian B, Ahmad N, Hunter CJ, Karver MR, Stupp SI, Sharma AK. Self‐Assembling Nanofibers Inhibit Inflammation in a Murine Model of Crohn's‐Disease‐Like Ileitis. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202000274] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Matthew I. Bury
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Natalie J. Fuller
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Tristan D. Clemons
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Department of Chemistry Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
| | - Renea M. Sturm
- Department of Urology University of California Los Angeles 200 Medical Plaza Driveway #140 Los Angeles CA 90095 USA
| | - Christopher D. Morrison
- Department of Urology Northwestern University Feinberg School of Medicine 676 North St. Clair Suite 2300 Chicago IL 60611 USA
| | - Devon C. Lisy‐Snow
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Bonnie G. Nolan
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Christopher Tarczynski
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
| | - Emily M. T. Ayello
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
| | - Amber Boyce
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Bridget Muckian
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Nida Ahmad
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Catherine J. Hunter
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Lurie Children's Hospital of Chicago 255 East Superior Street Chicago IL 60611 USA
| | - Mark R. Karver
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
| | - Samuel I. Stupp
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Department of Chemistry Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
- McCormick School of Engineering Department of Biomedical Engineering Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
- Department of Materials Science and Engineering Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
- Feinberg School of Medicine Department of Medicine Northwestern University 420 E Superior Street Chicago IL 60611 USA
| | - Arun K. Sharma
- Simpson Querrey Institute (SQI) Northwestern University 303 East Superior Street Chicago IL 60611 USA
- Department of Urology Northwestern University Feinberg School of Medicine 676 North St. Clair Suite 2300 Chicago IL 60611 USA
- McCormick School of Engineering Department of Biomedical Engineering Northwestern University 2145 Sheridan Road Evanston IL 60208 USA
- Stanley Manne Children's Research Institute (SMCRI) 303 East Superior Street Chicago IL 60611 USA
- Center for Advanced Regenerative Engineering (CARE) 2145 Sheridan Road Evanston IL 60208 USA
| |
Collapse
|
11
|
Dziąbowska-Grabias K, Sztanke M, Zając P, Celejewski M, Kurek K, Szkutnicki S, Korga P, Bulikowski W, Sztanke K. Antioxidant Therapy in Inflammatory Bowel Diseases. Antioxidants (Basel) 2021; 10:antiox10030412. [PMID: 33803138 PMCID: PMC8000291 DOI: 10.3390/antiox10030412] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 02/24/2021] [Accepted: 03/01/2021] [Indexed: 12/18/2022] Open
Abstract
Inflammatory bowel diseases (IBD) are a group of chronic, incurable diseases of the digestive tract, the etiology of which remains unclear to this day. IBD result in significant repercussions on the quality of patients’ life. There is a continuous increase in the incidence and prevalence of IBD worldwide, and it is becoming a significant public health burden. Pharmaceuticals commonly used in IBD management, for example, mesalamine, sulfasalazine, corticosteroids, and others, expose patients to diverse, potentially detrimental side effects and frequently do not provide sufficient disease control. The chronic inflammation underlies the etiology of IBD and closely associates with oxidative/nitrosative stress and a vast generation of reactive oxygen/nitrogen species. Relative to this, several substances with antioxidant and anti-inflammatory properties are now intensively researched as possible adjunctive or independent treatment options in IBD. Representatives of several different groups, including natural and chemical compounds will be characterized in this dissertation.
Collapse
Affiliation(s)
- Katarzyna Dziąbowska-Grabias
- Department of Gastroenterology, 1st Military Research Hospital, and Polyclinic of Lublin, 20-049 Lublin, Poland; (K.D.-G.); (P.Z.); (M.C.)
| | - Małgorzata Sztanke
- Department of Medical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland
- Correspondence: ; Tel.: +48-814-486-195
| | - Przemysław Zając
- Department of Gastroenterology, 1st Military Research Hospital, and Polyclinic of Lublin, 20-049 Lublin, Poland; (K.D.-G.); (P.Z.); (M.C.)
| | - Michał Celejewski
- Department of Gastroenterology, 1st Military Research Hospital, and Polyclinic of Lublin, 20-049 Lublin, Poland; (K.D.-G.); (P.Z.); (M.C.)
| | - Katarzyna Kurek
- Department of Pneumonology, Oncology, and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (K.K.); (S.S.)
| | - Stanisław Szkutnicki
- Department of Pneumonology, Oncology, and Allergology, Medical University of Lublin, 20-090 Lublin, Poland; (K.K.); (S.S.)
| | - Patryk Korga
- Department of Gastroenterology, 10ft Military Research Hospital, and Polyclinic of Bydgoszcz, 85-681 Bydgoszcz, Poland;
| | | | - Krzysztof Sztanke
- Laboratory of Bioorganic Synthesis and Analysis, Chair and Department of Medical Chemistry, Medical University of Lublin, 20-093 Lublin, Poland;
| |
Collapse
|
12
|
Kristensen LB, Lambertsen KL, Nguyen N, Byg KE, Nielsen HH. The Role of Non-Selective TNF Inhibitors in Demyelinating Events. Brain Sci 2021; 11:brainsci11010038. [PMID: 33401396 PMCID: PMC7824660 DOI: 10.3390/brainsci11010038] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Revised: 12/21/2020] [Accepted: 12/22/2020] [Indexed: 12/31/2022] Open
Abstract
The use of non-selective tumor necrosis factor (TNF) inhibitors is well known in the treatment of inflammatory diseases such as rheumatoid arthritis, Crohn’s disease, and psoriasis. Its use in neurological disorders is limited however, due to rare adverse events of demyelination, even in patients without preceding demyelinating disease. We review here the molecular and cellular aspects of this neuroinflammatory process in light of a case of severe monophasic demyelination caused by treatment with infliximab. Focusing on the role of TNF, we review the links between CNS inflammation, demyelination, and neurodegenerative changes leading to permanent neurological deficits in a young woman, and we discuss the growing evidence for selective soluble TNF inhibitors as a new treatment approach in inflammatory and neurological diseases.
Collapse
Affiliation(s)
- Line Buch Kristensen
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, 5000 Odense C, Denmark; (L.B.K.); (K.L.L.); (K.-E.B.)
| | - Kate Lykke Lambertsen
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, 5000 Odense C, Denmark; (L.B.K.); (K.L.L.); (K.-E.B.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, St., 5000 Odense C, Denmark
- BRIDGE—Brain Research—Inter Disciplinary Guided Excellence, Department of Clinical Research, J.B. Winsloewsvej 19, 5000 Odense C, Denmark
| | - Nina Nguyen
- Department of Radiology, Odense University Hospital, J.B. Winsloewsvej 4, 5000 Odense C, Denmark;
| | - Keld-Erik Byg
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, 5000 Odense C, Denmark; (L.B.K.); (K.L.L.); (K.-E.B.)
- BRIDGE—Brain Research—Inter Disciplinary Guided Excellence, Department of Clinical Research, J.B. Winsloewsvej 19, 5000 Odense C, Denmark
- Rheumatology Research Unit, Odense University Hospital and University of Southern Denmark, Odense University Hospital, J.B. Winsloewsvej 4, 5000 Odense C, Denmark
| | - Helle H Nielsen
- Department of Neurology, Odense University Hospital, J.B. Winsloewsvej 4, 5000 Odense C, Denmark; (L.B.K.); (K.L.L.); (K.-E.B.)
- Department of Neurobiology Research, Institute of Molecular Medicine, University of Southern Denmark, J.B. Winsloewsvej 21, St., 5000 Odense C, Denmark
- BRIDGE—Brain Research—Inter Disciplinary Guided Excellence, Department of Clinical Research, J.B. Winsloewsvej 19, 5000 Odense C, Denmark
- Correspondence:
| |
Collapse
|
13
|
Nicolela Susanna F, Pavesio C. A review of ocular adverse events of biological anti-TNF drugs. J Ophthalmic Inflamm Infect 2020; 10:11. [PMID: 32337619 PMCID: PMC7184065 DOI: 10.1186/s12348-020-00202-6] [Citation(s) in RCA: 43] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 03/23/2020] [Indexed: 12/13/2022] Open
Abstract
The recent introduction of biological agents has revolutionized the treatment of chronic immune-inflammatory diseases; however, this new therapy did not come without significant side effects.Through large controlled studies indicating decrease in the number of uveitis flares, the role of TNF inhibitors therapy for non-infectious uveitis gained more ground. Paradoxically to its therapeutic effect, there are reports associating these drugs with the onset or recurrence of inflammatory eye disease.A number of studies have suggested possible roles for anti-TNF-α agents in precipitating or worsening an underlying inflammatory process, including the hypothesis of a disequilibrium in cytokine balance, but to date the mechanisms responsible for these adverse events are not fully understood.A PubMed literature search was performed using the following terms: ophthalmic complication, uveitis, inflammatory eye disease, optic neuritis, neuropathy, adverse events, anti-TNF, TNF alpha inhibitor, infliximab, etanercept, adalimumab, golimumab, certolizumab, and biologics. The data presented in this study was mainly derived from the use of TNF inhibitors in rheumatology, essentially because these drugs have been used for a longer period in this medical field.Many of the ocular adverse events reported on this review may be considered a paradoxical effect of anti-TNF therapy. We found a variety of data associating new onset of uveitis with anti-TNF therapy for rheumatic conditions, predominantly under etanercept.In conclusion, although there is increasing data on ocular adverse events, it remains to be seen whether the suggested link between TNF inhibitors and the onset of ocular inflammation is substantiated by more quality data. Nevertheless, the awareness of potential treatment side effects with anti-TNF should be highlighted.
Collapse
Affiliation(s)
| | - Carlos Pavesio
- Moorfields Eye Hospital NHS Foundation Trust and UCL Institute of Ophthalmology, London, UK
| |
Collapse
|
14
|
Mashayekhi K, Ganji A, Sankian M. Designing a new dimerized anti human TNF-α aptamer with blocking activity. Biotechnol Prog 2020; 36:e2969. [PMID: 31989789 DOI: 10.1002/btpr.2969] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 01/16/2020] [Accepted: 01/17/2020] [Indexed: 01/10/2023]
Abstract
The human tumor necrosis factor α (hTNF-α) is an important pro-inflammatory cytokine which plays critical roles in inflammatory diseases such as rheumatoid arthritis (RA). The anti-TNF-α proteins can reduce symptoms of RA. Due to limitations of protein-based therapies, it is necessary to find new anti-TNF-α agents instead of common anti-TNF-α proteins. Therefore, the aim of the current study was to identify a new DNA aptamer with anti-hTNF-α activity. The protein systematic evolution of ligands by exponential enrichment (SELEX) process was used for identifying DNA aptamers. Anti-hTNF-α aptamers were selected using dot blot, real-time PCR, and in vitro inhibitory assay. The selected aptamers were truncated in two steps, and finally, a dimer aptamer was constructed from different selected truncates to improve their inhibitory effect. Also, Etanercept was used as a positive control to inhibit TNF-α, in comparison to the designed aptamers. After 11 rounds, four aptamers with anti-hTNF-α inhibitory effect were identified. The truncation and dimerization strategy revealed a new dimer aptamer with 67 nM Kd , which has 40% inhibitory effect compared with Etanercept (60%). Overall, the dimerization and truncation aptamers could improve its activity. With regard to the several limitations of anti-TNF-α proteins therapies including immunogenicity, side effects, and cost-intensive, a new designed anti-hTNF-α dimer aptamer could be considered as a potential therapeutic and/or diagnostic agent for hTNF-α-related disorders.
Collapse
Affiliation(s)
- Kazem Mashayekhi
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali Ganji
- Department of Microbiology and Immunology, School of Medicine, Molecular and Medicine Research Center, Arak University of Medical Sciences, Arak, Iran
| | - Mojtaba Sankian
- Immuno-Biochemistry Lab, Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
15
|
Brenner P, Citarella A, Wingård L, Sundström A. Use of antidepressants and benzodiazepine-related hypnotics before and after initiation of TNF-α inhibitors or non-biological systemic treatment in patients with rheumatoid arthritis, psoriatic arthritis or ankylosing spondylitis. BMC Rheumatol 2020; 4:9. [PMID: 32072134 PMCID: PMC7014636 DOI: 10.1186/s41927-019-0106-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/18/2019] [Indexed: 12/27/2022] Open
Abstract
Background Rheumatoid arthritis (RA), psoriatic arthritis (PsA) and ankylosing spondylitis (AS) are autoimmune disorders associated with an increased risk for depression, anxiety and sleeping problems. The objective of this study was to analyze use of antidepressants and benzodiazepine-related hypnotics (BRH) in Sweden before and after first time treatment with anti-TNF and non-biological systemic (NBS) treatments among patients with the above diagnoses, and to correlate such use with that of randomly selected population controls. Methods Patients and dispensed drugs were identified in nationwide Swedish healthcare registers. Proportions of subjects filling prescriptions of antidepressants and BRH from 2 years before start of treatment (index-date), and 2 years after index date were assessed. Using the period -6 months to index-date as reference, prevalence rate ratios were computed for 6 months' intervals before and after index. For up to ten randomly selected population controls per patient, the same measures were calculated. Results A total of 6256 patients started anti-TNF treatment, and 13,241 NBS treatment. The mean age at index was 52.0 for the anti-TNF group and 56.1 for NBS. Use of antidepressants and BRH was similar in both treatment groups (10.4-12.8%), significantly more common than in the controls (6.6 to 7.6%). For all patients, proportions filling prescriptions for antidepressants and BRH decreased directly or soon after the index; no such changes were seen in the controls, who all showed a slow but steady increase in use over time. Starters of anti-TNF treatment did not show clearer decreases in use of psychotropics than those initiating NBS. Conclusions Decreased rates of dispensed psychotropic drugs after the time of anti-TNF and NBS treatment initiation were seen among patients with autoimmune disorders but not population controls. This may correspond to treatment effects of anti-TNF and NBS also on psychiatric symptoms among these patients.
Collapse
Affiliation(s)
- Philip Brenner
- Department of Medicine Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Anna Citarella
- Department of Medicine Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Louise Wingård
- Department of Medicine Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Anders Sundström
- Department of Medicine Solna, Centre for Pharmacoepidemiology, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| |
Collapse
|
16
|
Melsheimer R, Geldhof A, Apaolaza I, Schaible T. Remicade ® (infliximab): 20 years of contributions to science and medicine. Biologics 2019; 13:139-178. [PMID: 31440029 PMCID: PMC6679695 DOI: 10.2147/btt.s207246] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 05/16/2019] [Indexed: 12/17/2022]
Abstract
On August 24, 1998, Remicade® (infliximab), the first tumor necrosis factor-α (TNF) inhibitor, received its initial marketing approval from the US Food and Drug Administration for the treatment of Crohn’s disease. Subsequently, Remicade was approved in another five adult and two pediatric indications both in the USA and across the globe. In the 20 years since this first approval, Remicade has made several important contributions to the advancement of science and medicine: 1) clinical trials with Remicade established the proof of concept that targeted therapy can be effective in immune-mediated inflammatory diseases; 2) as the first monoclonal antibody approved for use in a chronic condition, Remicade helped in identifying methods of administering large, foreign proteins repeatedly while limiting the body’s immune response to them; 3) the need to establish Remicade’s safety profile required developing new methods and setting new standards for postmarketing safety studies, specifically in the real-world setting, in terms of approach, size, and duration of follow-up; 4) the study of Remicade has improved our understanding of TNF’s role in the immune system, as well as our understanding of the pathophysiology of a range of diseases characterized by chronic inflammation; and 5) Remicade and other TNF inhibitors have transformed treatment practices in these chronic inflammatory diseases: remission has become a realistic goal of therapy and long-term disability resulting from structural damage can be prevented. This paper reviews how, over the course of its development and 20 years of use in clinical practice, Remicade was able to make these contributions.
Collapse
Affiliation(s)
| | - Anja Geldhof
- Medical Affairs, Janssen Biologics BV, Leiden, the Netherlands
| | - Isabel Apaolaza
- Medical Affairs, Janssen Biologics BV, Leiden, the Netherlands
| | | |
Collapse
|
17
|
Flak MB, Colas RA, Muñoz-Atienza E, Curtis MA, Dalli J, Pitzalis C. Inflammatory arthritis disrupts gut resolution mechanisms, promoting barrier breakdown by Porphyromonas gingivalis. JCI Insight 2019; 4:125191. [PMID: 31292292 PMCID: PMC6629160 DOI: 10.1172/jci.insight.125191] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 05/22/2019] [Indexed: 12/11/2022] Open
Abstract
Rheumatoid arthritis is linked with altered host immune responses and severe joint destruction. Recent evidence suggests that loss of gut homeostasis and barrier breach by pathobionts, including Porphyromonas gingivalis, may influence disease severity. The mechanism(s) leading to altered gut homeostasis and barrier breakdown in inflammatory arthritis are poorly understood. In the present study, we found a significant reduction in intestinal concentrations of several proresolving mediators during inflammatory arthritis, including downregulation of the gut-protective mediator resolvin D5n-3 DPA (RvD5n-3 DPA). This was linked with increased metabolism of RvD5n-3 DPA to its inactive 17-oxo metabolite. We also found downregulation of IL-10 expression in the gut of arthritic mice that was coupled with a reduction in IL-10 and IL-10 receptor (IL-10R) in lamina propria macrophages. These changes were linked with a decrease in the number of mucus-producing goblet cells and tight junction molecule expression in the intestinal epithelium of arthritic mice when compared with naive mice. P. gingivalis inoculation further downregulated intestinal RvD5n-3 DPA and Il-10 levels and the expression of gut tight junction proteins. RvD5n-3 DPA, but not its metabolite 17-oxo-RvD5n-3 DPA, increased the expression of both IL-10 and IL-10R in macrophages via the upregulation of the aryl hydrocarbon receptor agonist l-kynurenine. Administration of RvD5n-3 DPA to arthritic P. gingivalis-inoculated mice increased intestinal Il-10 expression, restored gut barrier function, and reduced joint inflammation. Together, these findings uncover mechanisms in the pathogenesis of rheumatoid arthritis, where disruption of the gut RvD5n-3 DPA-IL-10 axis weakens the gut barrier, which becomes permissive to the pathogenic actions of the pathobiont P. gingivalis.
Collapse
Affiliation(s)
- Magdalena B. Flak
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London (QMUL), London, United Kingdom
| | - Romain A. Colas
- Lipid Mediator Unit, William Harvey Research Institute, QMUL, London, United Kingdom
| | - Estefanía Muñoz-Atienza
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London (QMUL), London, United Kingdom
| | | | - Jesmond Dalli
- Lipid Mediator Unit, William Harvey Research Institute, QMUL, London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, QMUL, London, United Kingdom
| | - Costantino Pitzalis
- Centre for Experimental Medicine & Rheumatology, William Harvey Research Institute, Queen Mary University of London (QMUL), London, United Kingdom
- Centre for Inflammation and Therapeutic Innovation, QMUL, London, United Kingdom
| |
Collapse
|
18
|
Delgado ME, Brunner T. The many faces of tumor necrosis factor signaling in the intestinal epithelium. Genes Immun 2019; 20:609-626. [DOI: 10.1038/s41435-019-0057-0] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 12/26/2018] [Indexed: 01/15/2023]
|
19
|
Berman S, Bucher J, Koyfman A, Long BJ. Emergent Complications of Rheumatoid Arthritis. J Emerg Med 2018; 55:647-658. [DOI: 10.1016/j.jemermed.2018.07.030] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2018] [Revised: 07/03/2018] [Accepted: 07/22/2018] [Indexed: 12/20/2022]
|
20
|
Pegoretti V, Baron W, Laman JD, Eisel ULM. Selective Modulation of TNF-TNFRs Signaling: Insights for Multiple Sclerosis Treatment. Front Immunol 2018; 9:925. [PMID: 29760711 PMCID: PMC5936749 DOI: 10.3389/fimmu.2018.00925] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Accepted: 04/13/2018] [Indexed: 12/26/2022] Open
Abstract
Autoimmunity develops when self-tolerance mechanisms are failing to protect healthy tissue. A sustained reaction to self is generated, which includes the generation of effector cells and molecules that destroy tissues. A way to restore this intrinsic tolerance is through immune modulation that aims at refurbishing this immunologically naïve or unresponsive state, thereby decreasing the aberrant immune reaction taking place. One major cytokine has been shown to play a pivotal role in several autoimmune diseases such as rheumatoid arthritis (RA) and multiple sclerosis (MS): tumor necrosis factor alpha (TNFα) modulates the induction and maintenance of an inflammatory process and it comes in two variants, soluble TNF (solTNF) and transmembrane bound TNF (tmTNF). tmTNF signals via TNFR1 and TNFR2, whereas solTNF signals mainly via TNFR1. TNFR1 is widely expressed and promotes mainly inflammation and apoptosis. Conversely, TNFR2 is restricted mainly to immune and endothelial cells and it is known to activate the pro-survival PI3K-Akt/PKB signaling pathway and to sustain regulatory T cells function. Anti-TNFα therapies are successfully used to treat diseases such as RA, colitis, and psoriasis. However, clinical studies with a non-selective inhibitor of TNFα in MS patients had to be halted due to exacerbation of clinical symptoms. One possible explanation for this failure is the non-selectivity of the treatment, which avoids TNFR2 stimulation and its immune and tissue protective properties. Thus, a receptor-selective modulation of TNFα signal pathways provides a novel therapeutic concept that might lead to new insights in MS pathology with major implications for its effective treatment.
Collapse
Affiliation(s)
- Valentina Pegoretti
- Department of Molecular Neurobiology (GELIFES), University of Groningen, Groningen, Netherlands
| | - Wia Baron
- Department of Cell Biology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Jon D Laman
- Department of Neuroscience, University Medical Center Groningen (UMCG), University of Groningen, Groningen, Netherlands
| | - Ulrich L M Eisel
- Department of Molecular Neurobiology (GELIFES), University of Groningen, Groningen, Netherlands
| |
Collapse
|
21
|
Kofoed MS, Fisker N, Christensen AE, Kjeldsen AD. Sinogenic intracranial complications: is adalimumab a culprit? BMJ Case Rep 2018; 2018:bcr-2017-221449. [PMID: 29348274 DOI: 10.1136/bcr-2017-221449] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
We present two 11-year-old girls with chronic recurrent multifocal osteomyelitis, treated with adalimumab. Both developed severe intracranial complications to sinusitis. Patient 1 had been treated with adalimumab for 15 months when she developed acute sinusitis complicated by an orbital abscess, forehead swelling, a subdural empyema and osteomyelitis of the frontal bone. She was treated with a rhinosurgical and neurosurgical approach with intravenous antibiotics.Patient 2 had been in adalimumab treatment for 10 weeks. Adalimumab was discontinued 8 weeks prior to developing subdural empyema and subcortical abscesses in combination with sinusitis. She was treated with endoscopic sinus surgery and intravenous antibiotics. Both patients had developed psoriasis and episodes of infection during treatment. They were non-septic and had low fever on presentation. None of the patients suffered any long-term neurological sequelae. The immunosuppressive treatment with adalimumab is considered to be the cause of the sinogenic intracranial complications in our cases.
Collapse
Affiliation(s)
| | - Niels Fisker
- Department of Paediatrics, The Hans Christian Andersen Children's Hospital, Odense, Denmark
| | | | | |
Collapse
|
22
|
Schneider A, Hossain I, VanderMolen J, Nicol K. Comparison of remicade to curcumin for the treatment of Crohn's disease: A systematic review. Complement Ther Med 2017; 33:32-38. [PMID: 28735823 DOI: 10.1016/j.ctim.2017.06.002] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2017] [Revised: 05/25/2017] [Accepted: 06/05/2017] [Indexed: 12/17/2022] Open
Abstract
OBJECTIVE The aim of this study was to review the literature to assess if there is evidence to support the use of Curcumin as a safe complementary therapy in treating Crohn's Disease (CD) in conjunction with Remicade. DESIGN Systematic searches were performed by three researchers using electronic databases (ProQuest Medical Library, CINAHL Complete, and PUBMED) to locate and identify articles to meet a predetermined set of inclusion criteria. Specifically full text, peer-reviewed articles published after 2007 were included if they studied human participants 18 years or older. RESULTS Tumor necrosis factor-alpha (TNF-a) and Interleukin-1 (IL-1) levels increase in CD patients. Remicade reduces TNF-a in adults with CD. The issues are eventual loss of response (LOR) once IL-1 increases, and severe risks such as malignancy. CD patients using Curcumin saw a 55 point mean reduction in the Crohn's Disease Activity Index, reducing IL-1 and Crp. Plus it reduced TNF-a and PPMTase which improved colorectal cancer outcomes. CONCLUSIONS LOR of Remicade occurs when IL-1 increases, and it can cause malignancy. Research shows Curcumin reduces IL-1 and improves cancer outcomes. Future research, using both Remicade and Curcumin, would have to be done, but preliminary data would suggest using both would reduce LOR. Curcumin, even by itself, was found to be a cheap and safe way to reduce CD symptoms and inflammatory markers.
Collapse
Affiliation(s)
- Allaire Schneider
- Grand Valley State University, 1 Campus Drive Allendale, MI 49401, United States
| | - Ivy Hossain
- Grand Valley State University, 1 Campus Drive Allendale, MI 49401, United States
| | - Julia VanderMolen
- Grand Valley State University, 515 Michigan, Suite 200 Office 205 Allendale, MI 49546, United States.
| | - Kara Nicol
- Grand Valley State University, 1 Campus Drive Allendale, MI 49401, United States
| |
Collapse
|
23
|
Chinthakindi PK, Singh J, Gupta S, Nargotra A, Mahajan P, Kaul A, Ahmed Z, Koul S, Sangwan PL. Synthesis of α-santonin derivatives for diminutive effect on T and B-cell proliferation and their structure activity relationships. Eur J Med Chem 2016; 127:1047-1058. [PMID: 27847171 DOI: 10.1016/j.ejmech.2016.11.018] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/24/2016] [Accepted: 11/08/2016] [Indexed: 11/18/2022]
Abstract
A new library of 20 compounds from α-santonin was synthesized and tested against Con-A induced T-cell proliferation and LPS-induced B-cell proliferation via MTT assay. The study resulted in the identification of potent immunosuppressant molecules, which were further screened along with α-santonin for Tumor Necrosis Factor Alpha (TNF-α) inhibitory activity. One of the molecules (7) at 10 μM showed equipotency to that of dexamethasone (1 μM conc.) used as a standard. Structure activity relationships of the synthesized compounds along with our earlier reported α-santonin derivatives have been studied. Inferences from the modifications carried out at all the three sites of α-santonin have been elaborated. Computational study of the active compounds shows TNF-α protein as its preferable target rather than Inosine Monophosphate Dehydrogenase (IMPDH).
Collapse
Affiliation(s)
- Praveen K Chinthakindi
- Bioorganic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Catalysis and Peptide Research Unit, University of KwaZulu-Natal, Durban, 4041, South Africa
| | - Jasvinder Singh
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India; Academy of Scientific and Innovative Research (AcSIR), CSIR-IIIM Campus, Jammu, India
| | - Shilpa Gupta
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Amit Nargotra
- Discovery Informatics, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Priya Mahajan
- Discovery Informatics, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Anupurna Kaul
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Zabeer Ahmed
- Cancer Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Surrinder Koul
- Bioorganic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| | - Payare L Sangwan
- Bioorganic Chemistry Division, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India.
| |
Collapse
|
24
|
Hoffman TW, van Kessel DA, van Velzen-Blad H, Grutters JC, Rijkers GT. Antibody replacement therapy in primary antibody deficiencies and iatrogenic hypogammaglobulinemia. Expert Rev Clin Immunol 2015; 11:921-33. [DOI: 10.1586/1744666x.2015.1049599] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
25
|
Abstract
BACKGROUND/AIMS Five million people currently live with Crohn's disease (CD) or ulcerative colitis, the two major forms of inflammatory bowel disease. Available treatments frequently result in side effects that compromise the immune health of the patient. Consequently, alternative therapies that cause fewer systemic effects are needed. Dioctahedral smectite clays have been utilized to treat medical conditions, including diarrheal and enteric disease. Herein, we report the ability of a refined dioctahedral smectite (NovaSil, NS) to sorb inflammatory proteins and reduce inflammation in a TNBS (2,4,6-trinitrobenzenesulfonic acid) mouse model of CD. We also investigated whether NS could rescue gut microbial diversity in TNBS-induced mice. METHODS ELISA, X-ray diffraction, and transmission electron microscopy were employed to characterize the NS-cytokine interaction in vitro. A TNBS mouse colitis model was utilized to study the efficacy of NS supplementation for 4 weeks. The three treatment groups included control, TNBS, and TNBS + NS. DNA was extracted from feces and sorted for bacterial phylogenetic analysis. RESULTS Results suggest that NS binds TNFα in vitro. In TNBS-treated mice, supplementation with NS significantly reduced weight loss, and serum proinflammatory cytokine levels (IL-2, IL-6, and IL-12, TNFα, IFNγ) compared with the TNBS group. TNBS-treated mice demonstrated a significant reduction in gut microbiota species richness when compared with the TNBS + NS group and control group. CONCLUSIONS NovaSil mitigated the effects of TNBS-induced colitis based on reduction in systemic markers of inflammation, significant improvement in weight gain, and intestinal microbial profile.
Collapse
|
26
|
Fujioka K, Kishida T, Ejima A, Yamamoto K, Fujii W, Murakami K, Seno T, Yamamoto A, Kohno M, Oda R, Yamamoto T, Fujiwara H, Kawahito Y, Mazda O. Inhibition of osteoclastogenesis by osteoblast-like cells genetically engineered to produce interleukin-10. Biochem Biophys Res Commun 2014; 456:785-91. [PMID: 25514036 DOI: 10.1016/j.bbrc.2014.12.040] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Accepted: 12/08/2014] [Indexed: 11/28/2022]
Abstract
Bone destruction at inflamed joints is an important complication associated with rheumatoid arthritis (RA). Interleukin-10 (IL-10) may suppress not only inflammation but also induction of osteoclasts that play key roles in the bone destruction. If IL-10-producing osteoblast-like cells are induced from patient somatic cells and transplanted back into the destructive bone lesion, such therapy may promote bone remodeling by the cooperative effects of IL-10 and osteoblasts. We transduced mouse fibroblasts with genes for IL-10 and Runx2 that is a crucial transcription factor for osteoblast differentiation. The IL-10-producing induced osteoblast-like cells (IL-10-iOBs) strongly expressed osteoblast-specific genes and massively produced bone matrix that were mineralized by calcium phosphate in vitro and in vivo. Culture supernatant of IL-10-iOBs significantly suppressed induction of osteoclast from RANKL-stimulated Raw264.7 cells as well as LPS-induced production of inflammatory cytokine by macrophages. The IL-10-iOBs may be applicable to novel cell-based therapy against bone destruction associated with RA.
Collapse
Affiliation(s)
- Kazuki Fujioka
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan; Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Tsunao Kishida
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Akika Ejima
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Kenta Yamamoto
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan; Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Wataru Fujii
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ken Murakami
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Takahiro Seno
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan; Department of Rheumatic Diseases and Joint Function, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Aihiro Yamamoto
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Masataka Kohno
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Ryo Oda
- Department of Orthopaedics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiro Yamamoto
- Department of Dental Medicine, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Hiroyoshi Fujiwara
- Department of Orthopaedics, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yutaka Kawahito
- Inflammation and Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Osam Mazda
- Department of Immunology, Kyoto Prefectural University of Medicine, Kyoto, Japan.
| |
Collapse
|
27
|
Loosbroock C, Hunter KW. Inhibiting TNF-α signaling does not attenuate induction of endotoxin tolerance. J Inflamm Res 2014; 7:159-67. [PMID: 25506235 PMCID: PMC4259568 DOI: 10.2147/jir.s75037] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Tumor necrosis factor-alpha (TNF-α) is a central mediator of inflammatory responses elicited by Toll-like receptor agonists, such as the Gram-negative bacterial outer membrane antigen lipopolysaccharide (LPS). TNF-α is responsible for altering vascular permeability and activating infiltrating inflammatory cells, such as monocytes and neutrophils. Interestingly, TNF-α has also demonstrated the ability to induce tolerance to subsequent challenges with TNF-α or LPS in monocyte and macrophage cell populations. Tolerance is characterized by the inability to mount a typical inflammatory response during subsequent challenges following the initial exposure to an inflammatory mediator such as LPS. The ability of TNF-α to induce a tolerant-like state with regard to LPS is most likely a regulatory mechanism to prevent excessive inflammation. We hypothesized that the induction of tolerance or the degree of tolerance is dependent upon the production of TNF-α during the primary response to LPS. To investigate TNF-α-dependent tolerance, human monocytic THP-1 cells were treated with TNF-α-neutralizing antibodies or antagonistic TNF-α receptor antibodies before primary LPS stimulation and then monitored for the production of TNF-α during the primary and challenge stimulation. During the primary stimulation, anti-TNF-α treatment effectively attenuated the production of TNF-α and interleukin-1β; however, this reduced production did not impact the induction of endotoxin tolerance. These results demonstrate that interfering with TNF-α signaling attenuates production of inflammatory cytokines without affecting the induction of tolerance.
Collapse
Affiliation(s)
- Christopher Loosbroock
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, USA
| | - Kenneth W Hunter
- Department of Microbiology and Immunology, University of Nevada School of Medicine, Reno, NV, USA
| |
Collapse
|
28
|
Zenker S, Panteleev-Ivlev J, Wirtz S, Kishimoto T, Waldner MJ, Ksionda O, Tybulewicz VLJ, Neurath MF, Atreya I. A key regulatory role for Vav1 in controlling lipopolysaccharide endotoxemia via macrophage-derived IL-6. THE JOURNAL OF IMMUNOLOGY 2014; 192:2830-2836. [PMID: 24532586 DOI: 10.4049/jimmunol.1300157] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Macrophages are centrally involved in the pathogenesis of acute inflammatory diseases, peritonitis, endotoxemia, and septic shock. However, the molecular mechanisms controlling such macrophage activation are incompletely understood. In this article, we provide evidence that Vav1, a member of the RhoGEF family, plays a crucial role in macrophage activation and septic endotoxemia. Vav1-deficient mice demonstrated a significantly increased susceptibility for LPS endotoxemia that could be abrogated by anti-IL-6R Ab treatment. Subsequent studies showed that Vav1-deficient macrophages display augmented production of the proinflammatory cytokine IL-6. Nuclear Vav1 was identified as a key negative regulator of macrophage-derived IL-6 production. In fact, Vav1 formed a nuclear DNA-binding complex with heat shock transcription factor 1 at the HSE2 region of the IL-6 promoter to suppress IL-6 gene transcription in macrophages. These findings provide new insights into the pathogenesis of endotoxemia and suggest new avenues for therapy.
Collapse
Affiliation(s)
- Stefanie Zenker
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Julia Panteleev-Ivlev
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Stefan Wirtz
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | | | - Maximilian J Waldner
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Olga Ksionda
- MRC National Institute for Medical Research, London, United Kingdom
| | | | - Markus F Neurath
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| | - Imke Atreya
- Medical Clinic 1, Friedrich-Alexander University of Erlangen-Nürnberg, University Hospital of Erlangen, Germany
| |
Collapse
|
29
|
Puig L, Carrascosa J, Carretero G, de la Cueva P, Lafuente-Urrez R, Belinchón I, Sánchez-Regaña M, García-Bustínduy M, Ribera M, Alsina M, Ferrándiz C, Fonseca E, García-Patos V, Herrera E, López-Estebaranz J, Marrón S, Moreno J, Notario J, Rivera R, Rodriguez-Cerdeira C, Romero A, Ruiz-Villaverde R, Taberner R, Vidal D. Spanish Evidence-Based Guidelines on the Treatment of Psoriasis With Biologic Agents, 2013. Part 1: On Efficacy and Choice of Treatment. ACTA ACUST UNITED AC 2013; 104:694-709. [DOI: 10.1016/j.adengl.2013.04.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2013] [Accepted: 04/02/2013] [Indexed: 02/08/2023]
|
30
|
Puig L, Carrascosa J, Carretero G, de la Cueva P, Lafuente-Urrez R, Belinchón I, Sánchez-Regaña M, García-Bustínduy M, Ribera M, Alsina M, Ferrándiz C, Fonseca E, García-Patos V, Herrera E, Estebaranz JL, Marrón S, Moreno J, Notario J, Rivera R, Rodriguez-Cerdeira C, Romero A, Ruiz-Villaverde R, Taberner R, Vidal D. Directrices españolas basadas en la evidencia para el tratamiento de la psoriasis con agentes biológicos, 2013. I. Consideraciones de eficacia y selección del tratamiento. ACTAS DERMO-SIFILIOGRAFICAS 2013. [DOI: 10.1016/j.ad.2013.04.003] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
|
31
|
Johnston SS, Turpcu A, Shi N, Fowler R, Chu BC, Alexander K. Risk of infections in rheumatoid arthritis patients switching from anti-TNF agents to rituximab, abatacept, or another anti-TNF agent, a retrospective administrative claims analysis. Semin Arthritis Rheum 2013; 43:39-47. [PMID: 23453683 DOI: 10.1016/j.semarthrit.2012.12.024] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2012] [Revised: 12/20/2012] [Accepted: 12/24/2012] [Indexed: 10/27/2022]
Abstract
OBJECTIVE This study compared the incidence and hazard of ICD-9-CM-coded infections and severe infections in rheumatoid arthritis (RA) patients treated with subsequent-line (SL) BIOs (BIO) after switching from first-line (FL) anti-TNF therapy (anti-TNF). METHODS Retrospective analysis of a large U.S. claims database. RA patients initiating an FL anti-TNF between 1/1/2004 and 3/31/2010 were identified and followed forward in time to capture all SL BIO episodes through 3/31/2010. SL BIO episodes were classified into: abatacept, adalimumab, etanercept, infliximab, or rituximab. Multivariate mixed-effects survival models compared the hazard of infections and severe infections across the SL BIO episodes with adjustment for demographic and clinical confounders. RESULTS In total, 4332 SL BIO episodes were identified: mean age 55 years; 80% female. In adjusted analyses: when compared to rituximab, the hazard of all infections was significantly higher for adalimumab (hazard ratio [HR] = 1.31, 95% confidence interval [CI] = 1.09-1.55), etanercept (HR = 1.44, 95% CI = 1.20-1.72), and infliximab (HR = 1.30, 95% CI = 1.07-1.57), and insignificantly different for abatacept (HR = 1.18, 95% CI = 0.98-1.41); when compared to rituximab, the hazard of severe infection was significantly higher for infliximab (HR = 1.62, 95% CI = 1.03-2.55), and insignificantly different for abatacept (HR = 1.21, 95% CI = 0.78-1.88), adalimumab (HR = 1.10, 95% CI = 0.72-1.68), and etanercept (HR = 1.27, 95% CI = 0.83-1.95). CONCLUSIONS In RA patients treated with SL BIO, a 30-44% higher hazard of all infection was observed in anti-TNFs versus rituximab with a 62% higher hazard of severe infection observed in infliximab versus rituximab. This study used a non-randomized, observational design and is therefore subject to confounding from unmeasured factors that influence both treatment choice and infection risk.
Collapse
Affiliation(s)
- Stephen S Johnston
- Outcomes Research, Truven Health Analytics, 7700 Old Georgetown Road, Bethesda, MD 20814, USA.
| | | | | | | | | | | |
Collapse
|
32
|
Luo Y, Pang Z, Zhu Q, Cai X, Yin Y, Wang M, Zhu J, Chen J, Zeng K, Zhang C, Zhang J. Locally instilled tumor necrosis factor-α antisense oligonucleotide inhibits allergic inflammation via the induction of Tregs. J Gene Med 2012; 14:374-83. [PMID: 22576979 DOI: 10.1002/jgm.2631] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Anti-tumor necrosis factor (TNF)-α therapeutics has the potential to alleviate allergic inflammation. However, in previous studies, the systemic administration of anti-TNF-α agents was frequently accompanied by many adverse effects, such as infection, immunogenicity and malignancy. Efforts are made in the present study to evaluate whether or not local administration of TNF-α antisense oligonucleotide would inhibit allergic airway inflammation and influence systemic immune responses in an ovalbumin-induced asthmatic murine model. METHODS The treatment effects of TNF-α antisense oligonucleotide on mice, as well as the alternative proportion of regulatory T cells and T(H) 2 cells, were examined and compared with untreated mice. RESULTS Local administration of TNF-α antisense oligonucleotide resulted in significantly inhibited TNF-α expression, remarkably decreased inflammatory cell infiltration and dramatically reduced mucus hypersecretion. These treatment effects were associated with induced CD4(+) CD25(+) Foxp3(+) regulatory T cells, reduced T(H) 2 cells and generally decreased T(H) 2-type cytokines expression in bronchoalveolar lavage fluid. Systemic immunosuppression was not triggered by local antisense oligonucleotide administration because the proportion of CD4(+) CD25(+) Foxp3(+) regulatory T cells in the blood, thymus or spleen was not affected. Attenuated 4-1BBL expression was likely involved in the alternative proportion of T cells. CONCLUSIONS These findings demonstrate that local administration of TNF-α antisense oligonucleotide contributes to anti-inflammatory action via the enhancement of regulatory T cells-mediated immune tolerance, which is not accompanied by systemic immunosuppression associated with systemically-induced regulatory T cells.
Collapse
Affiliation(s)
- Yi Luo
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Martin PL, Bugelski PJ. Concordance of preclinical and clinical pharmacology and toxicology of monoclonal antibodies and fusion proteins: soluble targets. Br J Pharmacol 2012; 166:806-22. [PMID: 22168335 DOI: 10.1111/j.1476-5381.2011.01812.x] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Monoclonal antibodies (mAbs) and fusion proteins directed towards soluble targets make an important contribution to the treatment of disease. The purpose of this review was to correlate the clinical and preclinical data on the 14 currently approved mAbs and fusion proteins targeted to soluble targets. The principal sources used to gather data were: the peer reviewed Literature; European Medicines Agency 'Scientific Discussions' and United States Food and Drug Administration 'Pharmacology/Toxicology Reviews' and package inserts (United States Prescribing Information). Data on the following approved biopharmaceuticals were included: adalimumab, anakinra, bevacizumab, canakinumab, certolizumab pegol, denosumab, eculizumab, etanercept, golimumab, infliximab, omalizumab, ranibizumab, rilonacept and ustekinumab. Some related biopharmaceuticals in late-stage development were also included for comparison. Good concordance with human pharmacodynamics was found for both non-human primates (NHPs) receiving the human biopharmaceutical and mice receiving rodent homologues (surrogates). In contrast, there was limited concordance for human adverse effects in genetically deficient mice, mice receiving surrogates or NHPs receiving the human pharmaceutical. In summary, the results of this survey show that although both mice and NHPs have good predictive value for human pharmacodynamics, neither species have good predictive value for human adverse effects. No evidence that NHPs have superior predictive value was found.
Collapse
Affiliation(s)
- Pauline L Martin
- Biologics Toxicology, Janssen Research & Development, Radnor, PA 19087, USA.
| | | |
Collapse
|
34
|
Katz J, Frank M. Prolonged fever after Infliximab infusion. World J Gastrointest Pharmacol Ther 2012; 3:34-5. [PMID: 22966481 PMCID: PMC3437439 DOI: 10.4292/wjgpt.v3.i3.34] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2011] [Revised: 01/31/2012] [Accepted: 02/08/2012] [Indexed: 02/06/2023] Open
Abstract
Pharmacologic management for ulcerative colitis (UC) has recently been expanded to include anti- tumor necrosis factor (TNF) therapy for severe disease. Infliximab, a chimeric monoclonal antibody directed again TNF α was first tested in patients with Crohn’s disease. In addition to serious infections, malignancy, drug induced lupus and other autoimmune diseases, serum sickness-like reactions, neurological disease, and infusion reactions further complicate the use of Infliximab. We report a case of prolonged fever after Infliximab infusion to treat steroid refractory UC.
Collapse
Affiliation(s)
- Jennifer Katz
- Jennifer Katz, Michael Frank, New York University Medical Center, New York, NY 10016, United States
| | | |
Collapse
|
35
|
Cohen M, Baldin B, Thomas P, Lebrun C. [Neurological adverse events under anti-TNF alpha therapy]. Rev Neurol (Paris) 2011; 168:33-9. [PMID: 22098827 DOI: 10.1016/j.neurol.2011.06.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 06/06/2011] [Accepted: 06/24/2011] [Indexed: 01/24/2023]
Abstract
INTRODUCTION Anti-TNF alpha treatments are increasingly prescribed in various rheumatological or gastroenterological inflammatory diseases. Several adverse events, including neurological episodes have been reported in the literature. Relation to treatment is a major concern and guidelines for management of those patients are not available. The aim of our study is to collect and analyze neurological adverse events occurring during anti-TNF alpha therapy, and to propose guidelines for diagnosis of demyelinating-induced diseases. METHODS All patients treated with anti-TNF alpha drug, who were addressed in our department following a neurological event, were collected. We gathered clinical data including previous neurological history and immunosuppressive treatments. Paraclinical data included brain and spinal MRI, CSF study and outcome after anti-TNF therapy was collected. RESULTS Nine patients were included in this study. Sex ratio was eight and mean age was 49±9 years. One patient had previous history of subarachnoïdian hemorrage. All the patients previously received immunosuppressive drugs, including methotrexate (nine) and leflunomide (four). Three patients had a brain MRI before initiation of anti-TNF treatment, which was normal. Clinical episode was stroke-like in three cases, clinically isolated syndrome (CIS) in five cases, and peripheral neuropathy in one case. MRI showed lesions suggestive of demyelinating T2 hyperintensities in four cases, vascular infarcts in two cases, and non-specific T2 hyperintensities in three cases. Barkhof and Tintore criteria were fulfilled in one of the four CIS cases. CSF study was available for six patients. It was normal (four cases), showed oligoclonal bands (one case) and lymphocytic meningitis (one case). Anti-TNF alpha discontinuation was decided in five cases. Outcome was favorable for eight patients. One patient, whom MRI fulfilled Barkhof and Tintore criteria, and CSF showed oligoclonal bands, further developed relapsing remitting multiple sclerosis. CONCLUSION Our study is compatible with data found in the literature. Barkhof and Tintore criteria and CSF study are useful in clinical practice to diagnose a first demyelinating event. Standardized paraclinical neurological explorations should be proposed to physicians who are in charge of anti-TNF treated patients.
Collapse
Affiliation(s)
- M Cohen
- Service de neurologie, hôpital Pasteur, CHU de Nice, 30 voie Romaine, BP 69, Nice cedex, France.
| | | | | | | |
Collapse
|
36
|
Eliyahu E, Wolfson T, Ge Y, Jepsen KJ, Schuchman EH, Simonaro CM. Anti-TNF-alpha therapy enhances the effects of enzyme replacement therapy in rats with mucopolysaccharidosis type VI. PLoS One 2011; 6:e22447. [PMID: 21887218 PMCID: PMC3159569 DOI: 10.1371/journal.pone.0022447] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2011] [Accepted: 06/22/2011] [Indexed: 11/19/2022] Open
Abstract
Background Although enzyme replacement therapy (ERT) is available for several lysosomal storage disorders, the benefit of this treatment to the skeletal system is very limited. Our previous work has shown the importance of the Toll-like receptor 4/TNF-alpha inflammatory pathway in the skeletal pathology of the mucopolysaccharidoses (MPS), and we therefore undertook a study to examine the additive benefit of combining anti-TNF-alpha therapy with ERT in a rat model of MPS type VI. Methodology/Principal Findings MPS VI rats were treated for 8 months with Naglazyme® (recombinant human N-acetyl-galactosamine-4-sulfatase), or by a combined protocol using Naglazyme® and the rat-specific anti-TNF-alpha drug, CNTO1081. Both protocols led to markedly reduced serum levels of TNF-alpha and RANKL, although only the combined treatment reduced TNF-alpha in the articular cartilage. Analysis of cultured articular chondrocytes showed that the combination therapy also restored collagen IIA1 expression, and reduced expression of the apoptotic marker, PARP. Motor activity and mobility were improved by ERT, and these were significantly enhanced by combination treatment. Tracheal deformities in the MPS VI animals were only improved by combination therapy, and there was a modest improvement in bone length. Ceramide levels in the trachea also were markedly reduced. MicroCT analysis did not demonstrate any significant positive effects on bone microarchitecture from either treatment, nor was there histological improvement in the bone growth plates. Conclusions/Significance The results demonstrate that combining ERT with anti-TNF- alpha therapy improved the treatment outcome and led to significant clinical benefit. They also further validate the usefulness of TNF-alpha, RANKL and other inflammatory molecules as biomarkers for the MPS disorders. Further evaluation of this combination approach in other MPS animal models and patients is warranted.
Collapse
Affiliation(s)
- Efrat Eliyahu
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Theodore Wolfson
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Yi Ge
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Karl J. Jepsen
- Department of Orthopaedics, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Edward H. Schuchman
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
| | - Calogera M. Simonaro
- Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York, United States of America
- * E-mail:
| |
Collapse
|