1
|
Recent advances in gout drugs. Eur J Med Chem 2022; 245:114890. [DOI: 10.1016/j.ejmech.2022.114890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 10/24/2022] [Accepted: 10/25/2022] [Indexed: 11/24/2022]
|
2
|
Jiang Q, Wang X, Huang E, Wang Q, Wen C, Yang G, Lu L, Cui D. Inflammasome and Its Therapeutic Targeting in Rheumatoid Arthritis. Front Immunol 2022; 12:816839. [PMID: 35095918 PMCID: PMC8794704 DOI: 10.3389/fimmu.2021.816839] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/21/2021] [Indexed: 12/30/2022] Open
Abstract
Inflammasome is a cytoplasmic multiprotein complex that facilitates the clearance of exogenous microorganisms or the recognition of endogenous danger signals, which is critically involved in innate inflammatory response. Excessive or abnormal activation of inflammasomes has been shown to contribute to the development of various diseases including autoimmune diseases, neurodegenerative changes, and cancers. Rheumatoid arthritis (RA) is a chronic and complex autoimmune disease, in which inflammasome activation plays a pivotal role in immune dysregulation and joint inflammation. This review summarizes recent findings on inflammasome activation and its effector mechanisms in the pathogenesis of RA and potential development of therapeutic targeting of inflammasome for the immunotherapy of RA.
Collapse
Affiliation(s)
- Qi Jiang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Xin Wang
- Department of Rheumatology and Immunology, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Enyu Huang
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Qiao Wang
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chengping Wen
- School of Basic Medical Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Guocan Yang
- Department of Blood Transfusion, Shaoxing People's Hospital (Shaoxing Hospital, Zhejiang University School of Medicine), Shaoxing, China
| | - Liwei Lu
- Department of Pathology and Shenzhen Institute of Research and Innovation, The University of Hong Kong, Hong Kong, Hong Kong SAR, China.,Chongqing International Institute for Immunology, Chongqing, China
| | - Dawei Cui
- Department of Blood Transfusion, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
3
|
Jiang C, Xie S, Yang G, Wang N. Spotlight on NLRP3 Inflammasome: Role in Pathogenesis and Therapies of Atherosclerosis. J Inflamm Res 2022; 14:7143-7172. [PMID: 34992411 PMCID: PMC8711145 DOI: 10.2147/jir.s344730] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 12/03/2021] [Indexed: 12/12/2022] Open
Abstract
Inflammation is an intricate biological response of body tissues to detrimental stimuli. Cardiovascular disease (CVD) is the leading cause of death worldwide, and inflammation is well documented to play a role in the development of CVD, especially atherosclerosis (AS). Emerging evidence suggests that activation of the NOD-like receptor (NLR) family and the pyridine-containing domain 3 (NLRP3) inflammasome is instrumental in inflammation and may result in AS. The NLRP3 inflammasome acts as a molecular platform that triggers the activation of caspase-1 and the cleavage of pro-interleukin (IL)-1β, pro-IL-18, and gasdermin D (GSDMD). The cleaved GSDMD forms pores in the cell membrane and initiates pyroptosis, inducing cell death and the discharge of intracellular pro-inflammatory factors. Hence, the NLRP3 inflammasome is a promising target for anti-inflammatory therapy against AS. In this review, we systematically summarized the current understanding of the activation mechanism of NLRP3 inflammasome, and the pathological changes in AS involving NLRP3. We also discussed potential therapeutic strategies targeting NLRP3 inflammasome to combat AS.
Collapse
Affiliation(s)
- Chunteng Jiang
- Department of Internal Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China.,Department of Cardiology and Pneumology, University Medical Center of Göttingen, Georg-August-University of Göttingen, Göttingen, Lower Saxony, Germany
| | - Santuan Xie
- Department of Internal Medicine, The Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, People's Republic of China
| | - Guang Yang
- Department of Food Nutrition and Safety, School of Public Health, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| | - Ningning Wang
- Department of Food Nutrition and Safety, School of Public Health, Dalian Medical University, Dalian, Liaoning, People's Republic of China
| |
Collapse
|
4
|
Nunes PR, Mattioli SV, Sandrim VC. NLRP3 Activation and Its Relationship to Endothelial Dysfunction and Oxidative Stress: Implications for Preeclampsia and Pharmacological Interventions. Cells 2021; 10:cells10112828. [PMID: 34831052 PMCID: PMC8616099 DOI: 10.3390/cells10112828] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 10/14/2021] [Accepted: 10/15/2021] [Indexed: 01/12/2023] Open
Abstract
Preeclampsia (PE) is a specific syndrome of human pregnancy, being one of the main causes of maternal death. Persistent inflammation in the endothelium stimulates the secretion of several inflammatory mediators, activating different signaling patterns. One of these mechanisms is related to NLRP3 activation, initiated by high levels of danger signals such as cholesterol, urate, and glucose, producing IL-1, IL-18, and cell death by pyroptosis. Furthermore, reactive oxygen species (ROS), act as an intermediate to activate NLRP3, contributing to subsequent inflammatory cascades and cell damage. Moreover, increased production of ROS may elevate nitric oxide (NO) catabolism and consequently decrease NO bioavailability. NO has many roles in immune responses, including the regulation of signaling cascades. At the site of inflammation, vascular endothelium is crucial in the regulation of systemic inflammation with important implications for homeostasis. In this review, we present the important role of NLRP3 activation in exacerbating oxidative stress and endothelial dysfunction. Considering that the causes related to these processes and inflammation in PE remain a challenge for clinical practice, the use of drugs related to inhibition of the NLRP3 may be a good option for future solutions for this disease.
Collapse
|
5
|
Seok JK, Kang HC, Cho YY, Lee HS, Lee JY. Therapeutic regulation of the NLRP3 inflammasome in chronic inflammatory diseases. Arch Pharm Res 2021; 44:16-35. [PMID: 33534121 PMCID: PMC7884371 DOI: 10.1007/s12272-021-01307-9] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2020] [Accepted: 01/07/2021] [Indexed: 12/13/2022]
Abstract
Inflammasomes are cytosolic pattern recognition receptors that recognize pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) derived from invading pathogens and damaged tissues, respectively. Upon activation, the inflammasome forms a complex containing a receptor protein, an adaptor, and an effector to induce the autocleavage and activation of procaspase-1 ultimately culminating in the maturation and secretion of IL-1β and IL-18 and pyroptosis. Inflammasome activation plays an important role in host immune responses to pathogen infections and tissue repair in response to cellular damage. The NLRP3 inflammasome is a well-characterized pattern recognition receptor and is well known for its critical role in the regulation of immunity and the development and progression of various inflammatory diseases. In this review, we summarize recent efforts to develop therapeutic applications targeting the NLRP3 inflammasome to cure and prevent chronic inflammatory diseases. This review extensively discusses NLRP3 inflammasome-related diseases and current development of small molecule inhibitors providing beneficial information on the design of therapeutic strategies for NLRP3 inflammasome-related diseases. Additionally, small molecule inhibitors are classified depending on direct or indirect targeting mechanism to describe the current status of the development of pharmacological inhibitors.
Collapse
Affiliation(s)
- Jin Kyung Seok
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Han Chang Kang
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Yong-Yeon Cho
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Hye Suk Lee
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea
| | - Joo Young Lee
- BK21 PLUS Team, College of Pharmacy, The Catholic University of Korea, Bucheon, 14662, Republic of Korea.
| |
Collapse
|
6
|
Li Z, Guo J, Bi L. Role of the NLRP3 inflammasome in autoimmune diseases. Biomed Pharmacother 2020; 130:110542. [PMID: 32738636 DOI: 10.1016/j.biopha.2020.110542] [Citation(s) in RCA: 164] [Impact Index Per Article: 32.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/13/2020] [Accepted: 07/20/2020] [Indexed: 12/12/2022] Open
Abstract
NOD-like receptor family pyrin domain containing 3 (NLRP3) is an intracellular receptor that senses foreign pathogens and endogenous danger signals. It assembles with apoptosis-associated speck-like protein containing a CARD (ASC) and caspase-1 to form a multimeric protein called the NLRP3 inflammasome. Among its various functions, the NLRP3 inflammasome can induce the release of the pro-inflammatory cytokines interleukin (IL)-1β and IL-18 while also promoting gasdermin D (GSDMD)-mediated pyroptosis. Previous studies have established a vital role for the NLRP3 inflammasome in innate and adaptive immune system as well as its contribution to several autoimmune diseases including rheumatoid arthritis (RA), systemic lupus erythematosus (SLE), Sjögren's syndrome (SS), systemic sclerosis (SSc), and ankylosing spondylitis (AS). In this review, we briefly introduce the biological features of the NLRP3 inflammasome and present the mechanisms underlying its activation and regulation. We also summarize recent studies that have reported on the roles of NLRP3 inflammasome in the immune system and several autoimmune diseases, with a focus on therapeutic and clinical applications.
Collapse
Affiliation(s)
- Zhe Li
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Jialong Guo
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Liqi Bi
- Department of Rheumatology and Immunology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
7
|
Minimizing efficacy differences between phase II and III RCTs. Nat Rev Rheumatol 2020; 16:359-360. [DOI: 10.1038/s41584-020-0446-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
8
|
Rothwell R, Nikolov NP, Maynard JW, Levin G. Noninferiority Trials to Evaluate Drug Effects in Rheumatoid Arthritis. Arthritis Rheumatol 2020; 72:1258-1265. [PMID: 32182406 DOI: 10.1002/art.41257] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Accepted: 03/10/2020] [Indexed: 12/19/2022]
Abstract
OBJECTIVE The increased availability of highly effective treatments in rheumatoid arthritis (RA) necessitates a reexamination of study designs evaluating new treatments. We undertook this study to discuss possible specifications and considerations of noninferiority (NI) trials assessing drug effects in RA. METHODS We focused on the use of approved tumor necrosis factor inhibitors (TNFi) as potential active controls and reviewed previous placebo-controlled studies. We summarized the similarities in baseline characteristics and study design of the historical placebo-controlled studies used. After performing meta-analyses to estimate the effects of TNFi on symptoms, physical function, and radiographic progression in RA, we proposed NI margins and evaluated the feasibility of NI trials in this therapeutic setting. RESULTS We determined that an NI trial comparing an experimental treatment to a TNFi using the symptomatic end point of the American College of Rheumatology 20% improvement criteria response can feasibly provide evidence of a treatment effect, with a 12% absolute difference as one possible appropriate NI margin. For change from baseline in the Health Assessment Questionnaire disability index score, reasonable margins range from 0.10 to 0.12. In evaluating radiographic progression, an appropriate margin and the corresponding feasibility of the trial are dependent on the selected active control and the expected variability in progression. CONCLUSION Active-controlled studies in RA with justified NI margins can provide persuasive evidence of treatment effects on symptomatic, functional, and radiographic end points. Such studies can also provide reliable, controlled safety data and relevant information for treatment decisions in clinical practice. Thus, we recommend considering NI designs in future clinical trials in RA.
Collapse
|
9
|
Nagai K, Matsubayashi K, Ide K, Seto K, Kawasaki Y, Kawakami K. Factors Influencing Placebo Responses in Rheumatoid Arthritis Clinical Trials: A Meta-Analysis of Randomized, Double-Blind, Placebo-Controlled Studies. Clin Drug Investig 2020; 40:197-209. [PMID: 31953723 DOI: 10.1007/s40261-020-00887-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND AND OBJECTIVE A better understanding of placebo responses and the specific factors influencing these outcomes is important for clinical trial design. We investigated the magnitude of placebo responses at 3 months and the potential factors influencing these outcomes in rheumatoid arthritis (RA) clinical trials. METHODS We conducted a systematic review of randomized placebo-controlled trials of pharmacological agents for RA identified from PubMed, Embase, and Cochrane Central Register of Controlled Trials databases. The primary placebo outcome was American College of Rheumatology 20% response rate (ACR20). Data were pooled with a random-effects model. Factors influencing placebo response were assessed by meta-regression analyses. Subgroup analyses were performed for studies conducted in non-Western countries only versus in Western countries (North America and/or Europe) only or both. RESULTS The meta-analysis included 88 studies comprising 8406 patients receiving a placebo. The pooled estimate of placebo ACR20 was 29.0% (range 10.0-46.2; 95% confidence interval 27.2-30.9). Placebo ACR20 was negatively associated with trials in non-Western (Asian) countries and patient populations showing an inadequate response to biological disease-modifying antirheumatic drugs (DMARDs) in the multivariable analysis, whereas it was positively associated with the year of publication. No background DMARD treatment was also a negative predictor (albeit statistically non-significant). In subgroup analyses of Western and multiregional studies, study population and publication year were significant factors. CONCLUSIONS Our meta-analysis suggests that study location, patient population, and a background DMARD treatment influence placebo ACR20. These along with placebo response temporal profiles have important implications for designing and interpreting RA clinical trials.
Collapse
Affiliation(s)
- Kota Nagai
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Eisai Co., Ltd., 4-6-10 Koishikawa, Bunkyo-ku, Tokyo, 112-8088, Japan
| | - Keisuke Matsubayashi
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Kazuki Ide
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, 606-8501, Japan
| | - Kahori Seto
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan
| | - Yohei Kawasaki
- Biostatistics Section, Clinical Research Center, Chiba University Hospital, 1-8-1 Inohana, Chuo-ku, Chiba-shi, Chiba, 260-8677, Japan
| | - Koji Kawakami
- Department of Pharmacoepidemiology, Graduate School of Medicine and Public Health, Kyoto University, Yoshida-Konoe-cho, Sakyo-ku, Kyoto, 606-8501, Japan.
- Center for the Promotion of Interdisciplinary Education and Research, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
10
|
Zahid A, Li B, Kombe AJK, Jin T, Tao J. Pharmacological Inhibitors of the NLRP3 Inflammasome. Front Immunol 2019; 10:2538. [PMID: 31749805 DOI: 10.3389/fimmu.2019.02538/full] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/14/2019] [Indexed: 05/24/2023] Open
Abstract
Inflammasomes play a crucial role in innate immunity by serving as signaling platforms which deal with a plethora of pathogenic products and cellular products associated with stress and damage. By far, the best studied and most characterized inflammasome is NLRP3 inflammasome, which consists of NLRP3 (nucleotide-binding domain leucine-rich repeat (NLR) and pyrin domain containing receptor 3), ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain), and procaspase-1. Activation of NLRP3 inflammasome is mediated by highly diverse stimuli. Upon activation, NLRP3 protein recruits the adapter ASC protein, which recruits the procaspase-1 resulting in its cleavage and activation, inducing the maturation, and secretion of inflammatory cytokines and pyroptosis. However, aberrant activation of the NLRP3 inflammasome is implicated in various diseases including diabetes, atherosclerosis, metabolic syndrome, cardiovascular, and neurodegenerative diseases; raising a tremendous clinical interest in exploring the potential inhibitors of NLRP3 inflammasome. Recent investigations have disclosed various inhibitors of the NLRP3 inflammasome pathway which were validated through in vitro studies and in vivo experiments in animal models of NLRP3-associated disorders. Some of these inhibitors directly target the NLRP3 protein whereas some are aimed at other components and products of the inflammasome. Direct targeting of NLRP3 protein can be a better choice because it can prevent off target immunosuppressive effects, thus restrain tissue destruction. This paper will review the various pharmacological inhibitors of the NLRP3 inflammasome and will also discuss their mechanism of action.
Collapse
Affiliation(s)
- Ayesha Zahid
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bofeng Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Arnaud John Kombe Kombe
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Molecular Cell Science, Shanghai, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
11
|
Zahid A, Li B, Kombe AJK, Jin T, Tao J. Pharmacological Inhibitors of the NLRP3 Inflammasome. Front Immunol 2019; 10:2538. [PMID: 31749805 PMCID: PMC6842943 DOI: 10.3389/fimmu.2019.02538] [Citation(s) in RCA: 502] [Impact Index Per Article: 83.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2019] [Accepted: 10/14/2019] [Indexed: 12/14/2022] Open
Abstract
Inflammasomes play a crucial role in innate immunity by serving as signaling platforms which deal with a plethora of pathogenic products and cellular products associated with stress and damage. By far, the best studied and most characterized inflammasome is NLRP3 inflammasome, which consists of NLRP3 (nucleotide-binding domain leucine-rich repeat (NLR) and pyrin domain containing receptor 3), ASC (apoptosis-associated speck-like protein containing a caspase recruitment domain), and procaspase-1. Activation of NLRP3 inflammasome is mediated by highly diverse stimuli. Upon activation, NLRP3 protein recruits the adapter ASC protein, which recruits the procaspase-1 resulting in its cleavage and activation, inducing the maturation, and secretion of inflammatory cytokines and pyroptosis. However, aberrant activation of the NLRP3 inflammasome is implicated in various diseases including diabetes, atherosclerosis, metabolic syndrome, cardiovascular, and neurodegenerative diseases; raising a tremendous clinical interest in exploring the potential inhibitors of NLRP3 inflammasome. Recent investigations have disclosed various inhibitors of the NLRP3 inflammasome pathway which were validated through in vitro studies and in vivo experiments in animal models of NLRP3-associated disorders. Some of these inhibitors directly target the NLRP3 protein whereas some are aimed at other components and products of the inflammasome. Direct targeting of NLRP3 protein can be a better choice because it can prevent off target immunosuppressive effects, thus restrain tissue destruction. This paper will review the various pharmacological inhibitors of the NLRP3 inflammasome and will also discuss their mechanism of action.
Collapse
Affiliation(s)
- Ayesha Zahid
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Bofeng Li
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Arnaud John Kombe Kombe
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- CAS Center for Excellence in Molecular Cell Science, Shanghai, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
12
|
Bechman K, Yates M, Norton S, Cope AP, Galloway JB. Placebo Response in Rheumatoid Arthritis Clinical Trials. J Rheumatol 2019; 47:28-34. [PMID: 31043548 DOI: 10.3899/jrheum.190008] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/06/2019] [Indexed: 12/29/2022]
Abstract
OBJECTIVE Understanding the placebo response is critical to interpreting treatment efficacy, particularly for agents with a ceiling to their therapeutic effect, where an increasing placebo response makes it harder to detect potential benefit. The objective of this study is to assess the change in placebo responses over time in rheumatoid arthritis (RA) randomized placebo-controlled trials (RCT) for drug licensing authorization. METHODS The Cochrane Controlled Trials Register database was searched to identify RCT of biological or targeted synthetic disease-modifying antirheumatic drugs (DMARD) in RA. Studies were excluded if patients were conventional synthetic DMARD (csDMARD)-naive, not receiving background csDMARD therapy, or were biologic experienced. Metaregression model was used to evaluate changes in American College of Rheumatology (ACR) 20, ACR50, and ACR70 treatment response over time. RESULTS There were 32 trials in total: anti-tumor necrosis factor therapy (n = 15), tocilizumab (n = 4), abatacept (n = 2), rituximab (n = 2), and Janus kinase inhibitors (n = 9). From 1999 to 2018, there was no significant trend in the age or sex of patients in the placebo arm. Disease duration, swollen joint count, and 28-joint count Disease Activity Score using erythrocyte sedimentation rate at baseline all significantly declined over time. There was a statistically significant increase in placebo ACR50 and ACR70 responses (ACR50 β = 0.41, 95% CI 0.09-0.74, p = 0.01; ACR70 β = 0.18, 95% CI 0.04-0.31, p = 0.01) that remained significant after controlling for potential confounders. CONCLUSION There has been a rise in the placebo response in RA clinical trials over the last 2 decades. Shifting RA phenotype, changes in trial design, and expectation bias are possible explanations for this phenomenon. This observation has important implications when evaluating newer novel agents against established therapies.
Collapse
Affiliation(s)
- Katie Bechman
- From the Department of Inflammation Biology, Academic Rheumatology, and Psychology Department, Institute of Psychiatry, King's College London, London, UK. .,K. Bechman, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; M. Yates, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; S. Norton, PhD, Psychology Department, Institute of Psychiatry, King's College London; A. Cope, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London; J.B. Galloway, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London.
| | - Mark Yates
- From the Department of Inflammation Biology, Academic Rheumatology, and Psychology Department, Institute of Psychiatry, King's College London, London, UK.,K. Bechman, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; M. Yates, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; S. Norton, PhD, Psychology Department, Institute of Psychiatry, King's College London; A. Cope, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London; J.B. Galloway, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London
| | - Sam Norton
- From the Department of Inflammation Biology, Academic Rheumatology, and Psychology Department, Institute of Psychiatry, King's College London, London, UK.,K. Bechman, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; M. Yates, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; S. Norton, PhD, Psychology Department, Institute of Psychiatry, King's College London; A. Cope, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London; J.B. Galloway, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London
| | - Andrew P Cope
- From the Department of Inflammation Biology, Academic Rheumatology, and Psychology Department, Institute of Psychiatry, King's College London, London, UK.,K. Bechman, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; M. Yates, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; S. Norton, PhD, Psychology Department, Institute of Psychiatry, King's College London; A. Cope, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London; J.B. Galloway, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London
| | - James B Galloway
- From the Department of Inflammation Biology, Academic Rheumatology, and Psychology Department, Institute of Psychiatry, King's College London, London, UK.,K. Bechman, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; M. Yates, MBCHB, BSC, Department of Inflammation Biology, Academic Rheumatology, King's College London; S. Norton, PhD, Psychology Department, Institute of Psychiatry, King's College London; A. Cope, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London; J.B. Galloway, PhD, Department of Inflammation Biology, Academic Rheumatology, King's College London
| |
Collapse
|
13
|
Peterfy C, DiCarlo J, Emery P, Genovese MC, Keystone EC, Taylor PC, Schlichting DE, Beattie SD, Luchi M, Macias W. MRI and Dose Selection in a Phase II Trial of Baricitinib with Conventional Synthetic Disease-modifying Antirheumatic Drugs in Rheumatoid Arthritis. J Rheumatol 2019; 46:887-895. [PMID: 30647190 DOI: 10.3899/jrheum.171469] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2019] [Indexed: 11/22/2022]
Abstract
OBJECTIVE Magnetic resonance imaging (MRI) was used in a phase IIb study of baricitinib in patients with RA to support dose selection for the phase III program. METHODS Three hundred one patients with active RA who were taking stable methotrexate were randomized 2:1:1:1:1 to placebo or once-daily baricitinib (1, 2, 4, or 8 mg) for up to 24 weeks. One hundred fifty-four patients with definitive radiographic erosion had MRI of the hand/wrist at baseline and at weeks 12 and 24. Two expert radiologists, blinded to treatment and visit order, scored images for synovitis, osteitis, bone erosion, and cartilage loss. Combined inflammation (osteitis + 3× synovitis score) and total joint damage (erosion + 2.5× cartilage loss score) scores were calculated. Treatment groups were compared using ANCOVA adjusting for baseline scores. RESULTS Mean changes from baseline to Week 12 for synovitis were -0.10, -1.50, and -1.60 for patients treated with placebo, baricitinib 4 mg, and baricitinib 8 mg, respectively (p = 0.003 vs placebo for baricitinib 4 and 8 mg). Mean changes for osteitis were 0.00, -3.20, and -2.10 (p = 0.001 vs placebo for baricitinib 4 mg and p = 0.037 for 8 mg), respectively. Mean changes for bone erosion were 0.90, 0.10, and 0.40 (p = 0.089 for 4 mg and p = 0.275 for 8 mg), respectively, in these treatment groups. CONCLUSION MRI findings in this subgroup of patients suggest suppression of synovitis, osteitis, and combined inflammation by baricitinib 4 and 8 mg. This corroborates previously demonstrated clinical efficacy of baricitinib and increases confidence that baricitinib 4 mg could reduce the radiographic progression in phase III studies. [Clinical trial registration number (www.ClinicalTrials.gov): NCT01185353].
Collapse
Affiliation(s)
- Charles Peterfy
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada. .,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co.
| | - Julie DiCarlo
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Paul Emery
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Mark C Genovese
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Edward C Keystone
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Peter C Taylor
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Doug E Schlichting
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Scott D Beattie
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - Monica Luchi
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| | - William Macias
- From Spire Sciences Inc., Boca Raton, Florida; Division of Immunology and Rheumatology, Stanford University, Stanford, California; Eli Lilly and Co., Indianapolis, Indiana; Incyte Corporation, Wilmington, Delaware, USA; The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds, Leeds; Botnar Research Centre, University of Oxford, Oxford, UK; Department of Rheumatology, University of Toronto, Toronto, Ontario, Canada.,C. Peterfy, MD, PhD, Spire Sciences Inc.; J. DiCarlo, PhD, Spire Sciences Inc.; P. Emery, MD, FRCP, The Leeds Institute of Rheumatic and Musculoskeletal Medicine, University of Leeds; M.C. Genovese, MD, Division of Immunology and Rheumatology, Stanford University; E.C. Keystone, MD, FRCP, Department of Rheumatology, University of Toronto; P.C. Taylor, MA, PhD, FRCP, Botnar Research Centre, University of Oxford; D.E. Schlichting, RN, PhD, Eli Lilly and Co.; S.D. Beattie, PhD, Eli Lilly and Co.; M. Luchi, MD, FACR, MBA, Incyte Corp.; W. Macias, MD, PhD, Eli Lilly and Co
| |
Collapse
|
14
|
Pickles T, Christensen R, Tam LS, Simon LS, Choy EH. Early phase and adaptive design clinical trials in rheumatoid arthritis: a systematic review of early phase trials. Rheumatol Adv Pract 2018; 2:rky045. [PMID: 31431982 PMCID: PMC6649924 DOI: 10.1093/rap/rky045] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2018] [Revised: 09/17/2018] [Indexed: 12/21/2022] Open
Abstract
Objective Adaptive designs can enable highly sophisticated and efficient early phase trials, but the clinical inference from these trials is surrounded by complexity, and currently there is a paucity but steadily increasing amount of use of these designs in all fields of medicine. We aim to review early phase trials in RA to discover those that have used adaptive designs and benchmark trial characteristics. Methods From an OVID search for journal articles reporting the results of early phase trials in rheumatology, 35 studies were found, with 9 subsequently excluded; 11 were added from manual searches and 19 from searching the references. Study characteristics were extracted from the 56 papers (describing 62 trials), including the number of arms, number of patients, the primary outcome and when it was measured. Result One early phase trial using an adaptive design was found. The benchmark early phase trial in RA is a phase II double-blinded randomized trial, with four arms (one control and three intervention), each with 34 patients, and ACR20 measured at 16 weeks as the primary outcome. Conclusion The one adaptive design reviewed here, and a simulation study found in the search, both indicate that adaptive designs can be applied to early phase trials in RA. We have described the benchmark, which the efficiency of early phase trials using an adaptive design needs to exceed. These efficient designs could drive down numbers required, time for data collection and thus cost. Changes have been suggested, but more needs to be done.
Collapse
Affiliation(s)
- Tim Pickles
- Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University.,Centre for Trials Research, College of Biomedical and Life Sciences, Cardiff University, Cardiff, UK
| | - Robin Christensen
- Musculoskeletal Statistics Unit, The Parker Institute, Bispebjerg and Frederiksberg Hospital, Copenhagen, Denmark
| | - Lai-Shan Tam
- Department of Medicine & Therapeutics, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | | | - Ernest H Choy
- Cardiff Regional Experimental Arthritis Treatment and Evaluation (CREATE) Centre, Division of Infection and Immunity, School of Medicine, College of Biomedical and Life Sciences, Cardiff University
| |
Collapse
|
15
|
Rodríguez PC, Prada DM, Moreno E, Aira LE, Molinero C, López AM, Gómez JA, Hernández IM, Martínez JP, Reyes Y, Milera JM, Hernández MV, Torres R, Avila Y, Barrese Y, Viada C, Montero E, Hernández P. The anti-CD6 antibody itolizumab provides clinical benefit without lymphopenia in rheumatoid arthritis patients: results from a 6-month, open-label Phase I clinical trial. Clin Exp Immunol 2017; 191:229-239. [PMID: 28963724 DOI: 10.1111/cei.13061] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2017] [Indexed: 12/13/2022] Open
Abstract
Itolizumab is a humanized anti-CD6 monoclonal antibody (mAb) that has previously shown encouraging results, in terms of safety and positive clinical effects, in a 6-week monotherapy clinical trial conducted in rheumatoid arthritis (RA) patients. The current Phase I study evaluated the safety and clinical response for a longer treatment of 12 itolizumab intravenous doses in subjects with active RA despite previous disease-modifying anti-rheumatic drug (DMARD) therapy. Twenty-one subjects were enrolled into four dosage groups (0·1, 0·2, 0·4 and 0·8 mg/kg). Efficacy end-points including American College of Rheumatology (ACR)20, ACR50 and ACR70 response rates and disease activity score in 28 joints (DAS28) were monitored at baseline and at specific time-points during a 10-week follow-up period. Itolizumab was well tolerated up to the highest tested dose. No related serious adverse events were reported and most adverse events were mild. Remarkably, itolizumab treatment did not produce lymphopenia and, therefore, was not associated with infections. All patients achieved a clinical response (ACR20) at least once during the study. Eleven subjects (55%) achieved at least a 20% improvement in ACR just 1 week after the first itolizumab administration. The clinical response was observed from the beginning of the treatment and was sustained during 24 weeks. The efficacy profile of this 12-week treatment was similar to that of the previous study (6-week treatment). These results reinforce the safety profile of itolizumab and provide further evidence on the clinical benefit from the use of this anti-CD6 mAb in RA patients.
Collapse
Affiliation(s)
- P C Rodríguez
- Division of Clinical Research, Center of Molecular Immunology, Havana, Cuba
| | - D M Prada
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - E Moreno
- Facultad de Ciencias Básicas, Universidad de Medellin, Medellin, Colombia
| | - L E Aira
- Division of Clinical Research, Center of Molecular Immunology, Havana, Cuba
| | - C Molinero
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - A M López
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - J A Gómez
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - I M Hernández
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - J P Martínez
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - Y Reyes
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - J M Milera
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - M V Hernández
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - R Torres
- Service for Rheumatology, 10 de Octubre Hospital, Havana, Cuba
| | - Y Avila
- Department of Clinical Trials, National Coordinating Center of Clinical Trials, Havana, Cuba
| | - Y Barrese
- Department of Clinical Trials, National Coordinating Center of Clinical Trials, Havana, Cuba
| | - C Viada
- Division of Clinical Research, Center of Molecular Immunology, Havana, Cuba
| | - E Montero
- Experimental Immunotherapy Department, Center of Molecular Immunology, Havana, Cuba
| | - P Hernández
- Division of Clinical Research, Center of Molecular Immunology, Havana, Cuba
| |
Collapse
|
16
|
Biologic therapies and infections in the daily practice of three Italian rheumatologic units: a prospective, observational study. Clin Rheumatol 2016; 36:251-260. [DOI: 10.1007/s10067-016-3444-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/28/2016] [Accepted: 10/01/2016] [Indexed: 12/19/2022]
|
17
|
Scott DL, Ibrahim F, Farewell V, O'Keeffe AG, Ma M, Walker D, Heslin M, Patel A, Kingsley G. Randomised controlled trial of tumour necrosis factor inhibitors against combination intensive therapy with conventional disease-modifying antirheumatic drugs in established rheumatoid arthritis: the TACIT trial and associated systematic reviews. Health Technol Assess 2015; 18:i-xxiv, 1-164. [PMID: 25351370 DOI: 10.3310/hta18660] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Rheumatoid arthritis (RA) is initially treated with methotrexate and other disease-modifying antirheumatic drugs (DMARDs). Active RA patients who fail such treatments can receive tumour necrosis factor inhibitors (TNFis), which are effective but expensive. OBJECTIVE We assessed whether or not combination DMARDs (cDMARDs) give equivalent clinical benefits at lower costs in RA patients eligible for TNFis. DESIGN An open-label, 12-month, pragmatic, randomised, multicentre, two-arm trial [Tumour necrosis factor inhibitors Against Combination Intensive Therapy (TACIT)] compared these treatment strategies. We then systematically reviewed all comparable published trials. SETTING The TACIT trial involved 24 English rheumatology clinics. PARTICIPANTS Active RA patients eligible for TNFis. INTERVENTIONS The TACIT trial compared cDMARDs with TNFis plus methotrexate or another DMARD; 6-month non-responders received (a) TNFis if in the cDMARD group; and (b) a second TNFi if in the TNFi group. MAIN OUTCOME MEASURES The Heath Assessment Questionnaire (HAQ) was the primary outcome measure. The European Quality of Life-5 Dimensions (EQ-5D), joint damage, Disease Activity Score for 28 Joints (DAS28), withdrawals and adverse effects were secondary outcome measures. Economic evaluation linked costs, HAQ changes and quality-adjusted life-years (QALYs). RESULTS In total, 432 patients were screened; 104 started on cDMARDs and 101 started on TNFis. The initial demographic and disease assessments were similar between the groups. In total, 16 patients were lost to follow-up (nine in the cDMARD group, seven in the TNFi group) and 42 discontinued their intervention but were followed up (23 in the cDMARD group and 19 in the TNFi group). Intention-to-treat analysis with multiple imputation methods used for missing data showed greater 12-month HAQ score reductions with initial cDMARDs than with initial TNFis [adjusted linear regression coefficient 0.15, 95% confidence interval (CI) -0.003 to 0.31; p = 0.046]. Increases in 12-month EQ-5D scores were greater with initial cDMARDs (adjusted linear regression coefficient -0.11, 95% CI -0.18 to -0.03; p = 0.009) whereas 6-month changes in HAQ and EQ-5D scores and 6- and 12-month changes in joint damage were similar between the initial cDMARD group and the initial TNFi group. Longitudinal analyses (adjusted general estimating equations) showed that the DAS28 was lower in the initial TNFi group in the first 6 months (coefficient -0.63, 95% CI -0.93 to -0.34; p < 0.001) but there were no differences between the groups in months 6-12. In total, 36 patients in the initial cDMARD group and 44 in the initial TNFi group achieved DAS28 remission. The onset of remission did not differ between groups (p = 0.085 on log-rank test). In total, 10 patients in the initial cDMARD group and 18 in the initial TNFi group experienced serious adverse events; stopping therapy because of toxicity occurred in 10 and six patients respectively. Economic evaluation showed that the cDMARD group had similar or better QALY outcomes than TNFi with significantly lower costs at 6 and 12 months. In the systematic reviews we identified 32 trials (including 20-1049 patients) on early RA and 19 trials (including 40-982 patients) on established RA that compared (1) cDMARDs with DMARD monotherapy; (2) TNFis/methotrexate with methotrexate monotherapy; and (3) cDMARDs with TNFis/methotrexate. They showed that cDMARDs and TNFis had similar efficacies and toxicities. CONCLUSIONS Active RA patients who have failed methotrexate and another DMARD achieve equivalent clinical benefits at a lower cost from starting cDMARDs or from starting TNFis (reserving TNFis for non-responders). Only a minority of patients achieve sustained remission with cDMARDs or TNFis; new strategies are needed to maximise the frequency of remission. TRIAL REGISTRATION Current Control Trials ISRCTN37438295. FUNDING This project was funded by the National Institute for Health Research Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 18, No. 66. See the NIHR Journals Library website for further project information.
Collapse
Affiliation(s)
- David L Scott
- Department of Rheumatology, King's College London School of Medicine, London, UK
| | - Fowzia Ibrahim
- Department of Rheumatology, King's College London School of Medicine, London, UK
| | - Vern Farewell
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | - Aidan G O'Keeffe
- MRC Biostatistics Unit, Cambridge Institute of Public Health, Cambridge, UK
| | - Margaret Ma
- Department of Rheumatology, King's College London School of Medicine, London, UK
| | - David Walker
- Musculoskeletal Unit, Freeman Hospital, Newcastle upon Tyne, UK
| | - Margaret Heslin
- Centre for the Economics of Mental and Physical Health, Institute of Psychiatry, King's College London, London, UK
| | - Anita Patel
- Centre for the Economics of Mental and Physical Health, Institute of Psychiatry, King's College London, London, UK
| | - Gabrielle Kingsley
- Department of Rheumatology, King's College London School of Medicine, London, UK
| |
Collapse
|
18
|
Curtis JR, Chastek B, Becker L, Quach C, Harrison DJ, Yun H, Joseph GJ, Collier DH. Cost and effectiveness of biologics for rheumatoid arthritis in a commercially insured population. J Manag Care Spec Pharm 2015; 21:318-29. [PMID: 25803765 PMCID: PMC10398240 DOI: 10.18553/jmcp.2015.21.4.318] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
BACKGROUND Administrative claims contain detailed medication, diagnosis, and procedure data, but the lack of clinical outcomes for rheumatoid arthritis (RA) historically has limited their use in comparative effectiveness research. A claims-based algorithm was developed and validated to estimate effectiveness for RA from data for adherence, dosing, and treatment modifications. OBJECTIVE To implement the claims-based algorithm in a U.S. managed care database to estimate biologic cost per effectively treated patient. METHODS The cohort included patients with RA aged 18-63 years in the Optum Research Database who initiated biologic treatment between January 2007 and December 2010 and were continuously enrolled 6 months before through 12 months after the first claim for the biologic (the index date). Patients were categorized as effectively treated by the claims-based algorithm if they met all of the following 6 criteria in the 12-month post-index period: (1) a medication possession ratio ≥ 80% for subcutaneous biologics, or at least as many infusions as specified in U.S. labeling for intravenous biologics; (2) no increase in biologic dose; (3) no switch in biologics; (4) no new nonbiologic disease-modifying antirheumatic drug; (5) no new or increased oral glucocorticoid treatment; and (6) no more than 1 glucocorticoid injection. Drug costs (all biologics) and administration costs (intravenous biologics) were obtained from allowed amounts on claims. Biologic cost per effectively treated patient was defined as total 1-year biologic cost divided by the number of patients categorized by the algorithm as effectively treated with that index biologic. Sensitivity analysis was conducted to examine the total health care costs per effectively treated patient during the first year of biologic therapy. RESULTS A total of 5,474 individuals were included in the analysis. The index biologic was categorized as effective by the algorithm for 28.9% of patients overall, including 30.6% for subcutaneous biologics and 22.1% for intravenous biologics. The index biologic was categorized as effective in the first year for 32.7% of etanercept (794/2,425), 32.3% of golimumab (40/124), 30.2% of abatacept (89/295), 27.7% of adalimumab (514/1,857), and 19.0% of infliximab (147/773) patients. Mean 1-year biologic cost per effectively treated patient, as defined in the algorithm, was lowest for etanercept ($43,935), followed by golimumab ($49,589), adalimumab ($52,752), abatacept ($62,300), and infliximab ($101,402). The rank order in the sensitivity analysis was the same, except for golimumab and etanercept. CONCLUSIONS Using a claims-based algorithm in a large commercial claims database, etanercept was the most effective and had the lowest biologic cost per effectively treated patient with RA.
Collapse
Affiliation(s)
- Jeffrey R Curtis
- University of Alabama, Faculty Office Tower 820, 510 20th St. South, Birmingham, AL 35294.
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Dossing A, Tarp S, Furst DE, Gluud C, Beyene J, Hansen BB, Bliddal H, Christensen R. Interpreting trial results following use of different intention-to-treat approaches for preventing attrition bias: a meta-epidemiological study protocol. BMJ Open 2014; 4:e005297. [PMID: 25260368 PMCID: PMC4179424 DOI: 10.1136/bmjopen-2014-005297] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
INTRODUCTION When participants drop out of randomised clinical trials, as frequently happens, the intention-to-treat (ITT) principle does not apply, potentially leading to attrition bias. Data lost from patient dropout/lack of follow-up are statistically addressed by imputing, a procedure prone to bias. Deviations from the original definition of ITT are referred to as modified intention-to-treat (mITT). As yet, the impact of the potential bias associated with mITT has not been assessed. Our objective is to investigate potential bias and disadvantages of performing mITT and evaluate possible concerns when executing different mITT approaches in meta-analyses. METHODS AND ANALYSIS Using meta-epidemiology on randomised trials considered less prone to bias (ie, good internal validity) and assessing biological or targeted agents in patients with rheumatoid arthritis, we will meta-analyse data from 10 biological and targeted drugs based on collections of trials that would correspond to 10 individual meta-analyses. ETHICS AND DISSEMINATION This study will enhance transparency for evaluating mITT treatment effects described in meta-analyses. The intended audience will include healthcare researchers, policymakers and clinicians. Results of the study will be disseminated by peer-review publication. PROTOCOL REGISTRATION In PROSPERO CRD42013006702, 11. December 2013.
Collapse
Affiliation(s)
- Anna Dossing
- Musculoskeletal Statistics Unit, Department of Rheumatology, The Parker Institute, Copenhagen University Hospitals, Bispebjerg and Frederiksberg, Denmark
- Department of Clinical Medicine, Faculty of Medical and Health Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Simon Tarp
- Musculoskeletal Statistics Unit, Department of Rheumatology, The Parker Institute, Copenhagen University Hospitals, Bispebjerg and Frederiksberg, Denmark
| | - Daniel E Furst
- University of California, Los Angeles, David Geffen School of Medicine, Los Angeles, USA
| | - Christian Gluud
- Copenhagen Trial Unit, Centre for Clinical Intervention Research, Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark
| | - Joseph Beyene
- Department of Clinical Epidemiology & Biostatistics, Faculty of Health Sciences, McMaster University, Hamilton, Ontario, Canada
| | - Bjarke B Hansen
- Musculoskeletal Statistics Unit, Department of Rheumatology, The Parker Institute, Copenhagen University Hospitals, Bispebjerg and Frederiksberg, Denmark
| | - Henning Bliddal
- Musculoskeletal Statistics Unit, Department of Rheumatology, The Parker Institute, Copenhagen University Hospitals, Bispebjerg and Frederiksberg, Denmark
| | - Robin Christensen
- Musculoskeletal Statistics Unit, Department of Rheumatology, The Parker Institute, Copenhagen University Hospitals, Bispebjerg and Frederiksberg, Denmark
| |
Collapse
|
20
|
Favalli EG, Pregnolato F, Biggioggero M, Meroni PL. The comparison of effects of biologic agents on rheumatoid arthritis damage progression is biased by period of enrolment: Data from a systematic review and meta-analysis. Semin Arthritis Rheum 2014; 43:730-7. [DOI: 10.1016/j.semarthrit.2013.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Revised: 10/27/2013] [Accepted: 11/07/2013] [Indexed: 10/26/2022]
|
21
|
A clinical exploratory study with itolizumab, an anti-CD6 monoclonal antibody, in patients with rheumatoid arthritis. RESULTS IN IMMUNOLOGY 2012; 2:204-11. [PMID: 24371585 DOI: 10.1016/j.rinim.2012.11.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/09/2012] [Revised: 11/01/2012] [Accepted: 11/14/2012] [Indexed: 11/21/2022]
Abstract
T cells are involved in the pathogenesis of rheumatoid arthritis (RA). CD6 is a co-stimulatory molecule, predominantly expressed on lymphocytes, that has been linked to autoreactive responses. The purpose of this study was to evaluate the safety, immunogenicity and preliminary efficacy of itolizumab, a humanized anti-CD6 monoclonal antibody, in patients with active rheumatoid arthritis. Fifteen patients were enrolled in a phase I, open-label, dose-finding study. Five cohorts of patients received a weekly antibody monotherapy with a dose-range from 0.1 to 0.8 mg/kg. Itolizumab showed a good safety profile, with no severe or serious adverse events reported so far. No signs or symptoms associated with immunosuppression were observed in the study. Objective clinical responses were achieved in more than 80% of patients after treatment completion, and these responses tend to be sustained afterwards. This clinical study constitutes the first evidence of the safety and positive clinical effect of a monotherapy using an anti-CD6 antibody in patients with rheumatoid arthritis.
Collapse
Key Words
- ACR, American College of Rheumatology
- AE, adverse events
- CD6
- CRP, C reactive protein
- DMARD, disease-modifying antirheumatic drug
- ESR, eritrosedimentation rate
- Exploratory study
- NSAIDs, nonsteroidal antiinflammatory drugs
- RA, rheumatoid arthritis
- RF, rheumatoid factor
- Rheumatoid arthritis
- SAE, serious adverse event.
- T lymphocyte
- iv, intravenous
- mAbs, monoclonal antibodies
Collapse
|
22
|
Choy E, Taylor P, McAuliffe S, Roberts K, Sargeant I. Variation in the use of biologics in the management of rheumatoid arthritis across the UK. Curr Med Res Opin 2012; 28:1733-41. [PMID: 22978773 DOI: 10.1185/03007995.2012.731388] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
OBJECTIVES Treatment options for rheumatoid arthritis (RA) include conventional synthetic disease-modifying antirheumatic drugs (sDMARDs) and newer biologic DMARDs (biologics). This study describes treatment patterns, adherence to guidance and outcomes at hospital/regional level in the UK. METHODS This was a retrospective cohort study of RA patients selected from six health regions and from four hospitals in each region, including at least one major teaching hospital and two to three district/general hospitals. Treatment with sDMARDs/biologics was examined between November 2009 and June 2010 in comparison with National Institute for Health and Clinical Excellence (NICE) guidance and in relation to disease activity. RESULTS NICE guidance focuses on initiation of therapy and recommends combination sDMARD/biologic therapy. Analysis of 588 patient records indicates that overall 23% of patients were receiving biologic monotherapy. NICE guidance recommends initiation of biologics in patients with active RA (DAS28 >5.1): average DAS28 score on initiation of biologic therapy was above six in all regions. Range of DAS28 improvement post-biologic therapy was similar across all regions. DAS28 scores were well recorded within patient records at baseline and 6 months but not at other time points. CONCLUSIONS Our data highlight that nearly a quarter of patients receiving biologic treatments are not receiving concomitant sDMARD therapy and that DAS28 is poorly documented in patient medical records other than when initiating biologics and at the 6-month review. Limitations of this study include the limited sample size, the retrospective rather than prospective nature of the audit and the use of medical records only rather than other records such as nursing and pharmacy data. Nevertheless, the reasons for our findings merit further investigation to ensure that optimum treatment regimes and long term outcomes are being achieved for patients.
Collapse
Affiliation(s)
- Ernest Choy
- Department of Medicine, Cardiff University School of Medicine, UK.
| | | | | | | | | |
Collapse
|
23
|
Tan YK, Conaghan PG. Imaging in rheumatoid arthritis. Best Pract Res Clin Rheumatol 2012; 25:569-84. [PMID: 22137925 DOI: 10.1016/j.berh.2011.10.002] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2011] [Accepted: 09/10/2011] [Indexed: 01/29/2023]
Abstract
The optimal management of rheumatoid arthritis (RA) requires tools that allow early and accurate disease diagnosis, prediction of poor prognosis and responsive monitoring of therapeutic outcomes. Conventional radiography has been widely used in both clinical and research settings to assess RA joint damage due to its feasibility, but it has limitations in early disease detection and difficulty distinguishing between active treatments in modern trials. Imaging modalities such as magnetic resonance imaging (MRI) and ultrasound (US) have the advantage of detecting both joint inflammation and damage and hence they can provide additional and unique information. This can be especially useful in the context of early and/or undifferentiated joint disease when detection of soft tissue and bone marrow abnormalities is desirable. This review focusses on the recent literature concerning modern imaging, and provides clinicians with an insight into the role of imaging in modern RA diagnosis, prognosis and monitoring.
Collapse
Affiliation(s)
- York Kiat Tan
- Division of Musculoskeletal Disease, University of Leeds, UK
| | | |
Collapse
|
24
|
Schmitz S, Adams R, Walsh CD, Barry M, FitzGerald O. A mixed treatment comparison of the efficacy of anti-TNF agents in rheumatoid arthritis for methotrexate non-responders demonstrates differences between treatments: a Bayesian approach. Ann Rheum Dis 2012; 71:225-30. [PMID: 21960560 DOI: 10.1136/annrheumdis-2011-200228] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
BACKGROUND A number of tumour necrosis factor α (TNFα) antagonists (anti-TNFα) are available to treat rheumatoid arthritis. All of these have demonstrated considerable efficacy in placebo controlled trials, but few head-to-head comparisons exist to date. This work's objective is to estimate the relative efficacy among licensed anti-TNFs in patients who have had an inadequate response to methotrexate (MTX). Different outcome measures are used to highlight the advantages of continuous measures in such analyses. METHODS A systematic review identified randomised controlled trials comparing the efficacy of licensed anti-TNFα agents with placebo at 24 weeks in patients who have had an inadequate response to MTX. Relative efficacy was estimated using Bayesian mixed treatment comparison (MTC) models. Three different outcome measures were used: RR of achieving an American College of Rheumatology (ACR) 20 and ACR50 response and the percentage improvement in Health Assessment Questionnaire (HAQ) score. RESULTS 16 published trials were included in the analysis. All anti-TNFs show considerably improved efficacy over placebo. The MTC results also provide evidence of some differences in efficacy of the TNFα antagonists. Etanercept appears superior to infliximab and golimumab, and certolizumab to infliximab and adalimumab. ACR results indicate improved efficacy of certolizumab over golimumab. On HAQ analysis, adalimumab, certolizumab, etanercept and golimumab appear superior to infliximab, and etanercept shows improved efficacy compared with adalimumab. CONCLUSIONS There are differences in efficacy among the TNFα antagonists. In a MTC, a continuous outcome measure has more strength to detect such differences than a binomial outcome measure because of its enhanced sensitivity to change.
Collapse
|
25
|
Cantley MD, Bartold PM, Fairlie DP, Rainsford KD, Haynes DR. Histone deacetylase inhibitors as suppressors of bone destruction in inflammatory diseases. ACTA ACUST UNITED AC 2011; 64:763-74. [PMID: 22571254 DOI: 10.1111/j.2042-7158.2011.01421.x] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
OBJECTIVES Despite progress in developing many new anti-inflammatory treatments in the last decade, there has been little progress in finding treatments for bone loss associated with inflammatory diseases, such as rheumatoid arthritis and periodontitis. For instance, treatment of rheumatic diseases with anti-tumour necrosis factor-alpha agents has been largely successful in reducing inflammation, but there have been varying reports regarding its effectiveness at inhibiting bone loss. In addition, there is often a delay in finding the appropriate anti-inflammatory therapy for individual patients, and some therapies, such as disease modifying drugs, take time to have an effect. In order to protect the bone, adjunct therapies targeting bone resorption are being developed. This review focuses on new treatments based on using histone deacetylase inhibitors (HDACi) to suppress bone loss in these chronic inflammatory diseases. KEY FINDINGS A number of selected HDACi have been shown to suppress bone resorption by osteoclasts in vitro and in animal models of chronic inflammatory diseases. Recent reports indicate that these small molecules, which can be administered orally, could protect the bone and might be used in combination with current anti-inflammatory treatments. SUMMARY HDACi do have potential to suppress bone destruction in chronic inflammatory diseases including periodontitis and rheumatoid arthritis.
Collapse
Affiliation(s)
- Melissa D Cantley
- Discipline of Anatomy and Pathology, School of Medical Sciences, University of Adelaide, Adelaide, SA, Australia
| | | | | | | | | |
Collapse
|
26
|
Tanaka Y, Suzuki M, Nakamura H, Toyoizumi S, Zwillich SH. Phase II study of tofacitinib (CP-690,550) combined with methotrexate in patients with rheumatoid arthritis and an inadequate response to methotrexate. Arthritis Care Res (Hoboken) 2011; 63:1150-8. [PMID: 21584942 DOI: 10.1002/acr.20494] [Citation(s) in RCA: 239] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
OBJECTIVE To compare the efficacy, safety, and tolerability of 4 doses of oral tofacitinib (CP-690,550) with placebo in Japanese patients with active rheumatoid arthritis (RA) receiving stable background methotrexate (MTX) who had an inadequate response to MTX alone. METHODS A total of 140 patients were randomized to receive tofacitinib 1, 3, 5, and 10 mg twice a day or placebo in this 12-week, phase II, double-blind study. All patients remained on background MTX. Efficacy and safety were assessed at weeks 1, 2, 4, 8, and 12. The primary efficacy end point was the American College of Rheumatology 20% improvement criteria (ACR20) response rate at week 12. RESULTS ACR20 response rates at week 12 were significant (P < 0.0001) for all tofacitinib treatment groups: 1 mg twice a day, 64.3%; 3 mg twice a day, 77.8%; 5 mg twice a day, 96.3%; and 10 mg twice a day, 80.8% versus placebo, 14.3%. A significant dose-response relationship for the ACR20 was observed (P < 0.0001). Low disease activity was achieved by 72.7% of patients with high baseline disease activity for tofacitinib 10 mg twice a day at week 12 (P < 0.0001). Significant improvements in the ACR50, ACR70, Health Assessment Questionnaire Disability Index, and Disease Activity Score 28-3 (C-reactive protein) were also reported. The most commonly reported adverse events (AEs) were nasopharyngitis (n = 13) and increased alanine aminotransferase (n = 12) and aspartate aminotransferase (n = 9) levels. These AEs were mild or moderate in severity. Serious AEs were reported by 5 patients. No deaths occurred. CONCLUSION In Japanese patients with active RA with an inadequate response to MTX, tofacitinib in combination with MTX over 12 weeks was efficacious and had a manageable safety profile.
Collapse
Affiliation(s)
- Yoshiya Tanaka
- First Department of Internal Medicine, School of Medicine, University of Occupational and Environmental Health Japan, Iseigaoka, Yahatanishi, Kitakyushu, Japan.
| | | | | | | | | | | |
Collapse
|
27
|
Furfaro N, Mease PJ. Interpreting clinical trial results for moderate-to-severe rheumatoid arthritis: practical applications for rheumatology healthcare providers. ACTA ACUST UNITED AC 2011; 23:479-92. [PMID: 21899643 DOI: 10.1111/j.1745-7599.2011.00665.x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
PURPOSE To provide a general overview of clinical trials and more specifically define measurements common to rheumatoid arthritis clinical trials for the purpose of providing a foundation for rheumatology healthcare providers to translate clinical trial findings into their clinical practice and enhance their patient education discussions. DATA SOURCES English-language publications cited in the MEDLINE database were used to develop the content of this review article. CONCLUSIONS The role of rheumatology healthcare providers has evolved to include numerous vital functions, such as expanding communication between specialists and primary care providers, patient education and counseling, assistance with coping strategies, monitoring response to therapy, and administration of therapy. Education regarding clinical trial design, rationale, and discussion of endpoints has not been strongly emphasized for rheumatology healthcare providers who are increasingly introduced to novel agents and need to assimilate findings from clinical trials into daily practice. IMPLICATIONS FOR PRACTICE Familiarity with the basics of clinical trial design and efficacy endpoints of new rheumatoid arthritis therapeutics, translation and application of that knowledge into daily practice, and the ability to explain this information with patients will further enhance the ability of the rheumatology healthcare provider to optimize care for their patients with rheumatoid arthritis.
Collapse
Affiliation(s)
- Nicole Furfaro
- Seattle Rheumatology Associates, Seattle, Washington 98104, USA.
| | | |
Collapse
|
28
|
Rahman MU, Buchanan J, Doyle MK, Hsia EC, Gathany T, Parasuraman S, Aletaha D, Matteson EL, Conaghan PG, Keystone E, van der Heijde D, Smolen JS. Changes in patient characteristics in anti-tumour necrosis factor clinical trials for rheumatoid arthritis: results of an analysis of the literature over the past 16 years. Ann Rheum Dis 2011; 70:1631-40. [PMID: 21708910 PMCID: PMC3147244 DOI: 10.1136/ard.2010.146043] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/01/2011] [Indexed: 02/07/2023]
Abstract
OBJECTIVE To evaluate changes in baseline patient characteristics and entry criteria of randomised, controlled studies of tumour necrosis factor alpha (TNFα) inhibitors in rheumatoid arthritis (RA) patients. METHODS A systematic literature review was performed using predefined inclusion criteria to identify randomised, double-blind, controlled trials that evaluated TNFα inhibitors in adult RA patients. Entry criteria and baseline clinical characteristics were evaluated over time for methotrexate-experienced and methotrexate-naive study populations. Enrolment start date for each trial was the time metric. The anchor time was the study with the earliest identifiable enrolment start date. RESULTS 44 primary publications (reporting the primary study endpoint) from 1993 to 2008 met the inclusion criteria. Enrolment start dates of August 1993 and May 1997 were identified as time anchors for the 37 methotrexate-experienced studies and the seven methotrexate-naive studies, respectively. In methotrexate-experienced trials, no significant change was observed over the years included in this study in any inclusion criteria (including swollen joint counts and C-reactive protein (CRP)), but a significant decrease over time was observed in the baseline swollen joint count, CRP and total Sharp or van der Heijde modified Sharp score, but not in baseline tender joint counts. In the methotrexate-naive studies, significant decreases over the years were observed in swollen joint and tender joint inclusion criteria, but not in baseline tender joint count, baseline CRP, CRP inclusion criteria or baseline total Sharp or van der Heijde modified Sharp score. CONCLUSION Inclusion criteria and baseline characteristics of RA patients enrolled in studies of TNFα inhibitors have changed, with more recent trials enrolling cohorts with lower disease activity, especially in methotrexate-experienced trials.
Collapse
Affiliation(s)
- Mahboob U Rahman
- Centocor R&D, Inc, Malvern, Pennsylvania, USA
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- *Pfizer, Inc, Collegeville, Pennsylvania, USA
| | - Jacqui Buchanan
- Johnson & Johnson Pharmaceutical Services, Malvern, Pennsylvania, USA
- *Buchanan Biotech Consulting, Mountain View, California, USA
| | - Mittie K Doyle
- Centocor R&D, Inc, Malvern, Pennsylvania, USA
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Elizabeth C Hsia
- Centocor R&D, Inc, Malvern, Pennsylvania, USA
- University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Timothy Gathany
- Johnson & Johnson Pharmaceutical Services, Malvern, Pennsylvania, USA
| | | | - Daniel Aletaha
- Medical University of Vienna and Hietzing Hospital, Vienna, Austria
| | | | - Philip G Conaghan
- NIHR Leeds Musculoskeletal Biomedical Research Unit, University of Leeds, Leeds, UK
| | | | | | - Josef S Smolen
- Medical University of Vienna and Hietzing Hospital, Vienna, Austria
| |
Collapse
|
29
|
KARSH JACOB, KEYSTONE EDWARDC, HARAOUI BOULOS, THORNE JCARTER, POPE JANETE, BYKERK VIVIANP, MAKSYMOWYCH WALTERP, ZUMMER MICHEL, BENSEN WILLIAMG, KRAISHI MAJEDM. Canadian Recommendations for Clinical Trials of Pharmacologic Interventions in Rheumatoid Arthritis: Inclusion Criteria and Study Design: Table 1. J Rheumatol 2011; 38:2095-104. [DOI: 10.3899/jrheum.110188] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective.Current clinical trial designs for pharmacologic interventions in rheumatoid arthritis (RA) do not reflect the innovations in RA diagnosis, treatment, and care in countries where new drugs are most often used. The objective of this project was to recommend revised entry criteria and other study design features for RA clinical trials.Methods.Recommendations were developed using a modified nominal group consensus method. Canadian Rheumatology Research Consortium (CRRC) members were polled to rank the greatest challenges to clinical trial recruitment in their practices. Initial recommendations were developed by an expert panel of rheumatology trialists and other experts. A scoping study methodology was then used to examine the evidence available to support or refute each initial recommendation. The potential influence of CRRC recommendations on primary outcomes in future trials was examined. Recommendations were finalized using a consensus process.Results.Recommendations for clinical trial inclusion criteria addressed measures of disease activity [Disease Activity Score 28 using erythrocyte sedimentation rate (DAS28-ESR) > 3.2 PLUS ≥ 3 tender joints using 28-joint count (TJC28) PLUS ≥ 3 swollen joint (SJC28) OR C-reactive protein (CRP) or ESR > upper limit of normal PLUS ≥ 3 TJC28 PLUS ≥ 3 SJC28], functional classification, disease classification and duration, and concomitant RA treatments. Additional recommendations regarding study design addressed rescue strategies and longterm extension.Conclusion.There is an urgent need to modify clinical trial inclusion criteria and other study design features to better reflect the current characteristics of people living with RA in the countries where the new drugs will be used.
Collapse
|
30
|
Gibofsky A, Palmer WR, Keystone EC, Schiff MH, Feng J, McCroskery P, Baumgartner SW, Markenson JA. Rheumatoid arthritis disease-modifying antirheumatic drug intervention and utilization study: safety and etanercept utilization analyses from the RADIUS 1 and RADIUS 2 registries. J Rheumatol 2010; 38:21-8. [PMID: 20952478 DOI: 10.3899/jrheum.100347] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
OBJECTIVE to report the rates of serious adverse events (SAE), serious infectious events (SIE), and events of medical interest (EMI) in patients receiving etanercept; to identify the risk factors for SAE, SIE, and EMI; and to report time to switching from etanercept therapy, reasons for switching, and time to restarting treatment with etanercept in patients with rheumatoid arthritis (RA) in US clinical practice. METHODS adults ≥ 18 years of age who fulfilled the 1987 American Rheumatism Association criteria for RA were eligible for enrollment in 2 prospective, 5-year, multicenter, observational registries. RADIUS 1 (Rheumatoid Arthritis DMARD Intervention and Utilization Study) enrolled patients with RA who required a change in treatment [either an addition or a switch of a biologic or nonbiologic disease-modifying antirheumatic drug (DMARD)]. In RADIUS 2, patients with RA were required to start etanercept therapy at entry. Patients were seen at a frequency determined by their rheumatologist. RADIUS 1 and RADIUS 2 were registered under the US National Institutes of Health ClinicalTrials.gov identifiers NCT00116714 and NCT00116727, respectively. RESULTS in these patients, SAE, SIE, and EMI occurred at rates comparable to those seen in clinical trials. No unexpected safety signals were observed. Rates for SAE, SIE, and EMI in etanercept-treated patients were comparable to rates observed in patients receiving methotrexate monotherapy and did not increase with greater exposure to etanercept therapy. CONCLUSION the RADIUS registries provide a better understanding of the safety of etanercept in patients with RA in the US practice setting.
Collapse
Affiliation(s)
- Allan Gibofsky
- Hospital for Special Surgery, 535 East 70th Street, New York, NY 10021, USA.
| | | | | | | | | | | | | | | |
Collapse
|
31
|
Olech E. MRI in rheumatoid arthritis clinical trials: expensive imaging techniques may ultimately save money. Expert Rev Clin Pharmacol 2009; 2:443-447. [DOI: 10.1586/ecp.09.34] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023]
|